1
|
Xiao Y, Chen X, Li W, Li X, Zhou W. Impact of ACEI/ARB use on the survival of hypertensive patients with cancer: A meta‑analysis. Oncol Lett 2024; 28:534. [PMID: 39290956 PMCID: PMC11406588 DOI: 10.3892/ol.2024.14667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 07/01/2024] [Indexed: 09/19/2024] Open
Abstract
Angiotensin-converting enzyme inhibitors (ACEIs) and angiotensin receptor blockers (ARBs) are commonly used antihypertensive drugs. However, the impact that the use of ACEI and ARB drugs will have on the survival of patients with hypertension and cancer is still unclear. Therefore, the present study aimed to investigate the effects of ACEI and ARB use on the survival of patients with cancer. The Embase, PubMed and Web of Science databases were used to systematically analyze the survival of hypertensive patients with cancer treated with ACEIs or ARBs. Hazard ratios (HRs) and 95% confidence intervals (CIs) were calculated to evaluate the association between ACEI and ARB use and patient survival. The relationship between the survival of patients with certain types of cancer and ACEI and ARB use was evaluated using the calculated HRs. Patients with ovarian, pancreatic, prostate, hepatocellular, lung, esophageal, gastric, colon, nasopharyngeal, head and neck tumors, gallbladder and rectal cancers that used ACEI and ARB analogs had significantly increased survival times, except for patients with breast cancer (HR, 1.04; 95% CI, 0.90-1.19; P<0.01) and uroepithelial carcinoma (HR, 1.15; 95% CI, 0.69-1.94; P<0.01), who had significantly decreased survival times, when compared with patients who did not use these drugs. Analysis of the relationship between the use of ACEIs or ARBs alone or in combination on the overall survival of hypertensive patients with cancer demonstrated that the use of ACEIs alone (HR, 1.00; 95% CI, 0.93-1.08; P<0.01) did not have a significant effect on the survival of these patients. By contrast, the survival time was increased in hypertensive patients with cancer who used either ARBs alone (HR, 0.89; 95% CI, 0.84-0.94; P<0.01) or a combination of ACEIs and ARBs (HR, 0.84; 95% CI, 0.78-0.91; P<0.01). The present meta-analysis demonstrated the potential effects of ACEI and ARB use on the overall survival of patients with cancer. Therefore, investigation of the underlying mechanisms of action of ACEIs and ARBs, as well as the identification of specific groups of patients who may benefit from these interventions, could potentially lead to novel therapeutic options and improve the prognosis of patients with cancer in the future.
Collapse
Affiliation(s)
- Yao Xiao
- Department of General Surgery, The Second Clinical Medical College, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| | - Xinlong Chen
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shanxi 710061, P.R. China
| | - Wancheng Li
- Department of General Surgery, The Second Clinical Medical College, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| | - Xin Li
- Department of General Surgery, The Second Clinical Medical College, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| | - Wence Zhou
- Department of General Surgery, The Second Clinical Medical College, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| |
Collapse
|
2
|
Zhang R, Yin H, Yang M, Liu J, Zhen D, Zhang Z. Advanced progress of the relationship between renin-angiotensin-aldosterone system inhibitors and cancers. J Hypertens 2024; 42:1862-1873. [PMID: 39248142 DOI: 10.1097/hjh.0000000000003836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 07/29/2024] [Indexed: 09/10/2024]
Abstract
Hypertension and cancers are the most common causes of death in humans, as well as common co-diseases among elderly population. Studies have shown that hypertension is associated with carcinogenesis. The renin-angiotensin-aldosterone system (RAAS) is a crucial regulatory system of blood pressure, fluid, and electrolyte homeostasis, which plays an essential role in the pathogenesis of hypertension, whose mechanism is relatively clear. Studies have indicated that RAAS also widely exists in cancer tissues of different systems, which can affect the risk of cancers by stimulating cancer angiogenesis, participating in cancer-related oxidative stress, and regulating cancer-related immunity. Therefore, inhibiting RAAS activity seems beneficial to decreasing the risk of cancers. As one of the most commonly used antihypertensive drugs, RAAS inhibitors have been widely used in clinical practice. However, the conclusions of clinical studies on the relationship between RAAS inhibitors and cancers are not entirely consistent, which has been widely concerned by clinicians. The latest findings suggest that while RAAS inhibitors may reduce the risk of digestive cancers, respiratory cancers, urological cancers, gynecological cancers, and skin cancers, ACEIs may increase the risk of lung cancer, endometrial cancer, basal cell carcinoma, and squamous cell carcinoma. This article comprehensively reviews animal experiments, clinical studies, and meta-analyses on the relationship between RAAS inhibitors and cancers, to provide references for related studies in the future.
Collapse
Affiliation(s)
- Ruixing Zhang
- The First Clinical Medical College, Lanzhou University
- Department of Heart Center
| | - Hongtao Yin
- Department of Endocrinology, The First Hospital of Lanzhou University, Chengguan District, Lanzhou, Gansu, China
| | - Mengdi Yang
- The First Clinical Medical College, Lanzhou University
| | - Jinjin Liu
- Department of Endocrinology, The First Hospital of Lanzhou University, Chengguan District, Lanzhou, Gansu, China
| | - Donghu Zhen
- Department of Endocrinology, The First Hospital of Lanzhou University, Chengguan District, Lanzhou, Gansu, China
| | | |
Collapse
|
3
|
Pawlonka J, Buchalska B, Buczma K, Borzuta H, Kamińska K, Cudnoch-Jędrzejewska A. Targeting the Renin-angiotensin-aldosterone System (RAAS) for Cardiovascular Protection and Enhanced Oncological Outcomes: Review. Curr Treat Options Oncol 2024:10.1007/s11864-024-01270-9. [PMID: 39422794 DOI: 10.1007/s11864-024-01270-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2024] [Indexed: 10/19/2024]
Abstract
OPINION STATEMENT The renin-angiotensin-aldosterone system (RAAS) is a crucial regulator of the cardiovascular system and a target for widely used therapeutic drugs. Dysregulation of RAAS, implicated in prevalent diseases like hypertension and heart failure, has recently gained attention in oncological contexts due to its role in tumor biology and cardiovascular toxicities (CVTs). Thus, RAAS inhibitors (RAASi) may be used as potential supplementary therapies in cancer treatment and CVT prevention. Oncological treatments have evolved significantly, impacting patient survival and safety profiles. However, they pose cardiovascular risks, necessitating strategies for mitigating adverse effects. The main drug classes used in oncology include anthracyclines, anti-HER2 therapies, immune checkpoint inhibitors (ICIs), and vascular endothelial growth factor (VEGF) signaling pathway inhibitors (VSPI). While effective against cancer, these drugs induce varying CVTs. RAASi adjunctive therapy shows promise in enhancing clinical outcomes and protecting the cardiovascular system. Understanding RAAS involvement in cancer and CVT can inform personalized treatment approaches and improve patient care.
Collapse
Affiliation(s)
- J Pawlonka
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland.
| | - B Buchalska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - K Buczma
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - H Borzuta
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - K Kamińska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - A Cudnoch-Jędrzejewska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
4
|
Skelton WP, Masur J, Thomas J, Fallah P, Jain RK, Ravi P, Mantia C, McGregor BA, Nuzzo PV, Adib E, Zarif TE, Preston MA, Clinton TN, Li R, Steele GS, Kassouf W, Freeman D, Pond GR, Jain RK, Sonpavde GP. Impact of Angiotensin Converting Enzyme Inhibitors on Pathologic Complete Response With Neoadjuvant Chemotherapy for Muscle Invasive Bladder Cancer. Clin Genitourin Cancer 2024; 22:102143. [PMID: 39032202 DOI: 10.1016/j.clgc.2024.102143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/12/2024] [Accepted: 06/15/2024] [Indexed: 07/22/2024]
Abstract
INTRODUCTION The renin-angiotensin system (RAS) has been demonstrated to modulate cell proliferation, desmoplasia, angiogenesis and immunosuppression. We examined the association of RAS inhibitors (RASi)-namely angiotensin converting enzyme inhibitors (ACEi) and angiotensin receptor blockers (ARB)-with neoadjuvant chemotherapy (NAC) for muscle-invasive bladder cancer (MIBC) preceding radical cystectomy (RC). PATIENTS AND METHODS We retrospectively investigated concurrent RASi use with NAC prior to RC in 302 patients with MIBC from 3 academic institutions. Outcomes included pathologic complete response (pCR) and overall survival (OS). Pathologic features, performance status (PS), clinical stage, type/number of cycles of NAC, and toxicities were collected. RESULTS Overall pCR rate was 26.2% and 5-year OS was 62%. Concurrent ACEi intake with NAC approached significance for association with pCR (odds ratio [OR] = 1.71; 95% CI, 0.94-3.11; P = .077). Patients with cT3/4N0-N1 disease receiving ACEi had higher pCR rates (30.8% vs. 17.7%, P = .056) than those not on ACEi. Female sex had a statistically significant favorable interaction for pCR with ACEi intake (P = .044). ACEi intake was not associated with OS, while pCR, PS and lower clinical stage were significantly associated with improved OS. CONCLUSION ACEi intake is potentially associated with increased pCR in patients with MIBC receiving NAC prior to RC, and this association is more pronounced in patients with higher clinical stage of disease at the initiation of therapy and female sex. Our data suggest the potential relevance of the RAS as a therapeutic target in aggressive MIBC.
Collapse
Affiliation(s)
- William Paul Skelton
- University of Virginia Comprehensive Cancer Center, Department of Oncology, Charlottesville, VA; H. Lee Moffitt Cancer Center and Research Institute, Department of Oncology, Tampa, FL
| | - Jack Masur
- University of Virginia Comprehensive Cancer Center, Department of Oncology, Charlottesville, VA
| | - Jonathan Thomas
- Dana-Farber Cancer Institute, Department of Oncology, Boston, MA
| | - Parvaneh Fallah
- McGill University, Department of Oncology, Montreal, Quebec, Canada
| | - Rohit K Jain
- H. Lee Moffitt Cancer Center and Research Institute, Department of Oncology, Tampa, FL
| | - Praful Ravi
- Dana-Farber Cancer Institute, Department of Oncology, Boston, MA
| | - Charlene Mantia
- Dana-Farber Cancer Institute, Department of Oncology, Boston, MA
| | | | | | - Elio Adib
- Dana-Farber Cancer Institute, Department of Oncology, Boston, MA
| | - Talal El Zarif
- Dana-Farber Cancer Institute, Department of Oncology, Boston, MA
| | - Mark A Preston
- Brigham and Women's Hospital, Department of Oncology, Boston, MA
| | | | - Roger Li
- H. Lee Moffitt Cancer Center and Research Institute, Department of Oncology, Tampa, FL
| | - Graeme S Steele
- Brigham and Women's Hospital, Department of Oncology, Boston, MA
| | - Wassim Kassouf
- McGill University, Department of Oncology, Montreal, Quebec, Canada
| | - Dory Freeman
- Dana-Farber Cancer Institute, Department of Oncology, Boston, MA
| | | | - Rakesh K Jain
- Massachusetts General Hospital, Department of Oncology, Boston, MA
| | - Guru P Sonpavde
- Dana-Farber Cancer Institute, Department of Oncology, Boston, MA; AdventHealth Cancer Institute and the University of Central Florida, Department of Oncology, Orlando, FL.
| |
Collapse
|
5
|
Quan W, Xu CS, Ma C, Chen X, Yu DH, Li ZY, Wang DW, Tang F, Wan GP, Wan J, Wang ZF, Li ZQ. Anti-tumor effects of telmisartan in glioma-astrocyte non-contact co-cultures: A critical role of astrocytic IL-6-mediated paracrine growth promotion. Int Immunopharmacol 2024; 139:112707. [PMID: 39032472 DOI: 10.1016/j.intimp.2024.112707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/22/2024] [Accepted: 07/15/2024] [Indexed: 07/23/2024]
Abstract
Telmisartan, an angiotensin II type 1 receptor (AT1R) blocker, exhibits broad anti-tumor activity. However, in vitro, anti-proliferative effects are shown at doses far beyond the therapeutic plasma concentration. Considering the role of tumor microenvironment in glioma progression, glioma-astrocyte co-cultures were employed to test the anti-tumor potential of low-dose telmisartan. When a high dose was required for a direct anti-proliferative effect on glioma cell lines, a low dose significantly inhibited glioma cell proliferation and migration in the co-culture system. Under co-culture conditions, upregulated IL-6 expression in astrocytes played a critical role in glioma progression. Silencing IL-6 in astrocytes or IL-6R in glioma cells reduced proliferation and migration. Telmisartan (5 μM) inhibited astrocytic IL-6 expression, and its anti-tumor effects were reversed by silencing IL-6 or IL-6R and inhibiting signal transducer and activator of transcription 3 (STAT3) activity in glioma cells. Moreover, the telmisartan-driven IL-6 downregulation was not imitated by losartan, an AT1R blocker with little capacity of peroxisome proliferator-activated receptor-gamma (PPARγ) activation, but was eliminated by a PPARγ antagonist, indicating that the anti-glioma effects of telmisartan rely on its PPARγ agonistic activity rather than AT1R blockade. This study highlights the importance of astrocytic IL-6-mediated paracrine signaling in glioma growth and the potential of telmisartan as an adjuvant therapy for patients with glioma, especially those with hypertension.
Collapse
Affiliation(s)
- Wei Quan
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Cheng-Shi Xu
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Chao Ma
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xi Chen
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Dong-Hu Yu
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zhi-Yu Li
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Dan-Wen Wang
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Feng Tang
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Gui-Ping Wan
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jing Wan
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Ze-Fen Wang
- Department of Physiology, Wuhan University School of Basic Medical Sciences, Wuhan, Hubei, China.
| | - Zhi-Qiang Li
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China; Hubei International Science and Technology Cooperation Base for Research and Clinical Techniques for Brain Glioma Diagnosis and Treatment, Hubei, China.
| |
Collapse
|
6
|
Camarda ND, Lu Q, Meola DM, Man JJ, Song Z, Travers RJ, Lopez KE, Powers SN, Papanastasiou M, DeRuff KC, Mullahoo J, Egri SB, Davison D, Sebastiani P, Eblen ST, Buchsbaum R, Huggins GS, London CA, Jaffe JD, Upshaw JN, Yang VK, Jaffe IZ. Identifying mitigating strategies for endothelial cell dysfunction and hypertension in response to VEGF receptor inhibitors. Clin Sci (Lond) 2024; 138:1131-1150. [PMID: 39282930 DOI: 10.1042/cs20240537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 07/27/2024] [Accepted: 08/28/2024] [Indexed: 10/02/2024]
Abstract
Vascular endothelial growth factor receptor inhibitors (VEGFRis) improve cancer survival but are associated with treatment-limiting hypertension, often attributed to endothelial cell (EC) dysfunction. Using phosphoproteomic profiling of VEGFRi-treated ECs, drugs were screened for mitigators of VEGFRi-induced EC dysfunction and validated in primary aortic ECs, mice, and canine cancer patients. VEGFRi treatment significantly raised systolic blood pressure (SBP) and increased markers of endothelial and renal dysfunction in mice and canine cancer patients. α-Adrenergic-antagonists were identified as drugs that most oppose the VEGFRi proteomic signature. Doxazosin, one such α-antagonist, prevented EC dysfunction in murine, canine, and human aortic ECs. In mice with sorafenib-induced-hypertension, doxazosin mitigated EC dysfunction but not hypertension or glomerular endotheliosis, while lisinopril mitigated hypertension and glomerular endotheliosis without impacting EC function. Hence, reversing EC dysfunction was insufficient to mitigate VEGFRi-induced-hypertension in this mouse model. Canine cancer patients with VEGFRi-induced-hypertension were randomized to doxazosin or lisinopril and both agents significantly decreased SBP. The canine clinical trial supports safety and efficacy of doxazosin and lisinopril as antihypertensives for VEGFRi-induced-hypertension and the potential of trials in canines with spontaneous cancer to accelerate translation. The overall findings demonstrate the utility of phosphoproteomics to identify EC-protective agents to mitigate cardio-oncology side effects.
Collapse
Affiliation(s)
- Nicholas D Camarda
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, U.S.A
- Genetics, Molecular, and Cellular Biology Program, Tufts Graduate School of Biomedical Sciences, Boston, MA, U.S.A
| | - Qing Lu
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, U.S.A
| | - Dawn M Meola
- Tufts Cummings School of Veterinary Medicine, North Grafton, MA, U.S.A
| | - Joshua J Man
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, U.S.A
- Genetics, Molecular, and Cellular Biology Program, Tufts Graduate School of Biomedical Sciences, Boston, MA, U.S.A
| | - Zeyuan Song
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, MA, U.S.A
| | - Richard J Travers
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, U.S.A
- Division of Hematology Oncology, Department of Medicine, Tufts Medical Center, Boston, MA, U.S.A
| | - Katherine E Lopez
- Tufts Cummings School of Veterinary Medicine, North Grafton, MA, U.S.A
| | - Sarah N Powers
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, U.S.A
| | | | | | | | | | | | - Paola Sebastiani
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, MA, U.S.A
| | - Scott T Eblen
- Department of Cell and Molecular Pharmacology, Medical University of South Carolina, Charleston, SC, U.S.A
| | - Rachel Buchsbaum
- Division of Hematology Oncology, Department of Medicine, Tufts Medical Center, Boston, MA, U.S.A
| | - Gordon S Huggins
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, U.S.A
- Division of Cardiology, Tufts Medical Center, Boston, MA, U.S.A
| | - Cheryl A London
- Tufts Cummings School of Veterinary Medicine, North Grafton, MA, U.S.A
| | | | - Jenica N Upshaw
- Division of Cardiology, Tufts Medical Center, Boston, MA, U.S.A
| | - Vicky K Yang
- Tufts Cummings School of Veterinary Medicine, North Grafton, MA, U.S.A
| | - Iris Z Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, U.S.A
- Genetics, Molecular, and Cellular Biology Program, Tufts Graduate School of Biomedical Sciences, Boston, MA, U.S.A
| |
Collapse
|
7
|
Yang HY, Kang MY, Kang CM, Lee WJ, Hwang HK. Correlation between Angiotensin Inhibitor Administration and Longer Survival in Patients Who Underwent Curative Resection for Pancreatic Cancer. Yonsei Med J 2024; 65:324-331. [PMID: 38804026 PMCID: PMC11130588 DOI: 10.3349/ymj.2023.0399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 12/31/2023] [Accepted: 01/08/2024] [Indexed: 05/29/2024] Open
Abstract
PURPOSE The microenvironment of pancreatic ductal adenocarcinoma (PDAC) with extensive desmoplastic stroma contributes to aggressive cancer behavior. Angiotensin system inhibitors (ASIs) reduce stromal fibrosis and are a promising therapeutic strategy. The purpose of this study was to examine how ASIs affected the oncological results of patients who had their PDAC removed. MATERIALS AND METHODS A retrospective assessment was conducted on the clinicopathological and survival data of patients who received curative resection for PDAC at Severance Hospital between January 2012 and December 2019. RESULTS A total of 410 participants (228 male and 182 female), with a median follow-up period of 12.8 months, were included in this study. Patients were divided into three groups, based on ASI use and history of hypertension: group 1, normotensive and never used ASI (n=210, 51.2%); group 2, ASI non-users with hypertension (n=50, 12.2%); and group 3, ASI users with hypertension (n=150, 36.6%). The three groups did not differ significantly in terms of age, sex, kind of operation, T and N stages, or adjuvant and neoadjuvant therapy. Moreover, there was no discernible difference in disease-free survival between those who used ASI and those who did not (p=0.636). The 5-year overall survival (OS) rates in groups 1, 2, and 3 were 52.6%, 32.3%, and 38.0%, respectively. However, the OS rate of ASI users was remarkably higher than that of non-users (p=0.016). CONCLUSION In patients with resected PDAC, ASI is linked to longer survival rates. Furthermore, for individuals with hypertension, ASI in conjunction with conventional chemotherapy may be an easy and successful treatment option.
Collapse
Affiliation(s)
- Hye Yeon Yang
- Department of Liver Transplantation and Hepatobiliary-Pancreatic Surgery, Ajou University School of Medicine, Suwon, Korea
- Department of Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Min Yu Kang
- Department of Hepatobiliary and Pancreatic Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Chang Moo Kang
- Department of Hepatobiliary and Pancreatic Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Woo Jung Lee
- Department of Hepatobiliary and Pancreatic Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Ho Kyoung Hwang
- Department of Hepatobiliary and Pancreatic Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
8
|
Moriyama S, Hieda M, Kisanuki M, Kawano S, Yokoyama T, Fukata M, Kusaba H, Maruyama T, Baba E, Akashi K, Fukuda H. Both New-Onset and Pre-Existing Hypertension Indicate Favorable Clinical Outcomes in Patients Treated With Anti-Vascular Endothelial Growth Factor Therapy. Circ J 2024; 88:217-225. [PMID: 36476830 DOI: 10.1253/circj.cj-22-0628] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2024]
Abstract
BACKGROUND Hypertension is a frequent adverse event caused by vascular endothelial growth factor signaling pathway (VSP) inhibitors. However, the impact of hypertension on clinical outcomes during VSP inhibitor therapy remains controversial. METHODS AND RESULTS We reviewed 3,460 cancer patients treated with VSP inhibitors from the LIFE Study database, comprising Japanese claims data between 2016 and 2020. Patients were stratified into 3 groups based on the timing of hypertension onset: (1) new-onset hypertension (n=569; hypertension developing after VSP inhibitor administration); (2) pre-existing hypertension (n=1,790); and (3) no hypertension (n=1,101). Time to treatment failure (TTF) was used as the primary endpoint as a surrogate for clinical outcomes. The median (interquartile range) TTF in the new-onset and pre-existing hypertension groups was 301 (133-567) and 170 (72-358) days, respectively, compared with 146 (70-309) days in the non-hypertensive group (P<0.001 among all groups). In an adjusted Cox proportional hazard model, new-onset (hazard ratio [HR] 0.58; 95% confidence interval [CI] 0.50-0.68; P<0.001) and pre-existing (HR 0.85; 95% CI 0.73-0.98; P=0.026) hypertension were independent factors for prolonged TTF. The TTF of new-onset hypertension was longer than that of pre-existing hypertension (HR 0.68; 95% CI 0.62-0.76; P<0.001). CONCLUSIONS This study highlighted that new-onset hypertension induced by VSP inhibitors was an independent factor for favorable clinical outcomes. Pre-existing hypertension before VSP inhibitor initiation was also a significant factor.
Collapse
Affiliation(s)
- Shohei Moriyama
- Department of Hematology, Oncology and Cardiovascular Medicine, Kyushu University Hospital
| | - Michinari Hieda
- Department of Hematology, Oncology and Cardiovascular Medicine, Kyushu University Hospital
| | - Megumi Kisanuki
- Department of Hematology, Oncology and Cardiovascular Medicine, Kyushu University Hospital
| | - Shotaro Kawano
- Division of Immunology and Rheumatology, Hamanomachi Hospital
| | - Taku Yokoyama
- Department of Hematology, Oncology and Cardiovascular Medicine, Kyushu University Hospital
| | - Mitsuhiro Fukata
- Department of Hematology, Oncology and Cardiovascular Medicine, Kyushu University Hospital
| | - Hitoshi Kusaba
- Department of Hematology, Oncology and Cardiovascular Medicine, Kyushu University Hospital
- Division of Oncology, Hamanomachi Hospital
| | - Toru Maruyama
- Department of Hematology, Oncology and Cardiovascular Medicine, Kyushu University Hospital
- Division of Cardiology, Haradoi Hospital
| | - Eishi Baba
- Department of Oncology and Social Medicine, Kyushu University
| | - Koichi Akashi
- Department of Hematology, Oncology and Cardiovascular Medicine, Kyushu University Hospital
| | - Haruhisa Fukuda
- Department of Health Care Administration and Management, Kyushu University
| |
Collapse
|
9
|
Larrinaga G, Valdivia A, Arrieta-Aguirre I, Solano-Iturri JD, Ugalde-Olano A, Loizaga-Iriarte A, Santos-Martín A, Pérez-Fernández A, Angulo JC, López JI. The Expression of Alamandine Receptor MrgD in Clear Cell Renal Cell Carcinoma Is Associated with a Worse Prognosis and Unfavorable Response to Antiangiogenic Therapy. Int J Mol Sci 2024; 25:1499. [PMID: 38338778 PMCID: PMC10855800 DOI: 10.3390/ijms25031499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
Renal cell carcinoma (RCC) ranks among the most prevalent malignancies in Western countries, marked by its notable heterogeneity, which contributes to an unpredictable clinical trajectory. The insufficiency of dependable biomarkers adds complexity to assessing this tumor progression. Imbalances of several components of the intrarenal renin-angiotensin system (iRAS) significantly impact patient prognoses and responses to first-line immunotherapies. In this study, we analyzed the immunohistochemical expression of the Mas-related G-protein-coupled receptor D (MrgD), which recognizes the novel RAS peptide alamandine (ALA), in a series of 87 clear cell renal cell (CCRCCs), 19 papillary (PRCC), 7 chromophobe (ChRCC) renal cell carcinomas, and 11 renal oncocytomas (RO). MrgD was expressed in all the renal tumor subtypes, with a higher mean staining intensity in the PRCCs, ChRCCs, and ROs. A high expression of MrgD at the tumor center and at the infiltrative front of CCRCC tissues was significantly associated with a high histological grade, large tumor diameter, local invasion, and locoregional node and distant metastasis. Patients with worse 5-year cancer-specific survival and a poorer response to antiangiogenic tyrosine-kinase inhibitors (TKIs) showed higher MrgD expression at the center of their primary tumors. These findings suggest a possible role of MrgD in renal carcinogenetic processes. Further studies are necessary to unveil its potential as a novel biomarker for CCRCC prognosis and response to frontline therapies.
Collapse
Affiliation(s)
- Gorka Larrinaga
- Department of Nursing, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain;
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
- Biobizkaia Health Research Institute, 48903 Barakaldo, Spain; (J.D.S.-I.); (A.U.-O.); (A.L.-I.); (A.S.-M.); (A.P.-F.); (J.I.L.)
| | - Asier Valdivia
- Department of Cellular Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain;
| | - Inés Arrieta-Aguirre
- Department of Nursing, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain;
| | - Jon Danel Solano-Iturri
- Biobizkaia Health Research Institute, 48903 Barakaldo, Spain; (J.D.S.-I.); (A.U.-O.); (A.L.-I.); (A.S.-M.); (A.P.-F.); (J.I.L.)
- Department of Pathology, Cruces University Hospital, 48903 Barakaldo, Spain
| | - Aitziber Ugalde-Olano
- Biobizkaia Health Research Institute, 48903 Barakaldo, Spain; (J.D.S.-I.); (A.U.-O.); (A.L.-I.); (A.S.-M.); (A.P.-F.); (J.I.L.)
- Department of Pathology, Basurto University Hospital, 48903 Barakaldo, Spain
| | - Ana Loizaga-Iriarte
- Biobizkaia Health Research Institute, 48903 Barakaldo, Spain; (J.D.S.-I.); (A.U.-O.); (A.L.-I.); (A.S.-M.); (A.P.-F.); (J.I.L.)
- Department of Urology, Basurto University Hospital, University of the Basque Country (UPV/EHU), 48013 Bilbao, Spain
| | - Aida Santos-Martín
- Biobizkaia Health Research Institute, 48903 Barakaldo, Spain; (J.D.S.-I.); (A.U.-O.); (A.L.-I.); (A.S.-M.); (A.P.-F.); (J.I.L.)
- Department of Urology, Basurto University Hospital, University of the Basque Country (UPV/EHU), 48013 Bilbao, Spain
| | - Amparo Pérez-Fernández
- Biobizkaia Health Research Institute, 48903 Barakaldo, Spain; (J.D.S.-I.); (A.U.-O.); (A.L.-I.); (A.S.-M.); (A.P.-F.); (J.I.L.)
- Department of Urology, Basurto University Hospital, University of the Basque Country (UPV/EHU), 48013 Bilbao, Spain
| | - Javier C. Angulo
- Clinical Department, Faculty of Medical Sciences, European University of Madrid, 28905 Getafe, Spain;
- Department of Urology, University Hospital of Getafe, 28907 Madrid, Spain
| | - José I. López
- Biobizkaia Health Research Institute, 48903 Barakaldo, Spain; (J.D.S.-I.); (A.U.-O.); (A.L.-I.); (A.S.-M.); (A.P.-F.); (J.I.L.)
| |
Collapse
|
10
|
Regulska K, Michalak M, Kolenda T, Kozłowska-Masłoń J, Guglas K, Stanisz B. Angiotensin-converting enzyme inhibitors for ovarian cancer? - a new adjuvant option or a silent trap. Rep Pract Oncol Radiother 2023; 28:551-564. [PMID: 37795232 PMCID: PMC10547424 DOI: 10.5603/rpor.a2023.0059] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 07/24/2023] [Indexed: 10/06/2023] Open
Abstract
Background Ovarian cancer is a huge therapeutic and financial problem for which approved treatments have already achieved their limit of efficiency. A cost-effective strategy to extend therapeutic options in this malignancy is drug repurposing aimed at overcoming chemoresistance. Here, angiotensin-converting enzyme inhibitors (ACE-I) are worth considering. Materials and methods We searched literature for publications supporting the idea of adjuvant application of ACE-Is in ovarian malignancy. Then, we searched The Cancer Genome Atlas databases for relevant alternations of gene expression patterns. We also performed in silico structure-activity relationship evaluation for predicting ACE-Is' cytotoxicity against ovarian cancer cell lines. Finally, we reviewed the potential obstacles in ACE-Is repurposing process. Results The alternation of angiotensin receptor expression in ovarian cancer translates into poorer patient survival. This confirms the participation of the renin-angiotensin system in ovarian carcinogenesis. In observational studies, ACE-Is were shown synergize with both, platinum-based chemotherapy as well as with antiangiogenic therapy. Consistently, our in silico simulation showed that ACE-Is are probably cytotoxic against ovarian cancer cells. However, the publications on their chemopreventive properties were inconclusive. In addition, some reports correlated ACE-Is use with increased general cancer incidence. We hypothesized that this effect could be associated with mutagenic nitrosamine formation in ACE-Is' pharmaceutical formulations, as was the case with angiotensin receptor blockers (ARBs) and other well-established pharmaceuticals. Conclusions Available data warrant further research into repositioning ACE-Is to ovarian cancer as chemosensitizers. Prior to this, however, a special research program is needed to detect possible genotoxic contaminants of ACE-Is.
Collapse
Affiliation(s)
- Katarzyna Regulska
- Pharmacy, Greater Poland Cancer Centre, Poznan, Poland
- Department of Clinical Pharmacy and Biopharmacy, Poznan University of Medical Sciences, Poznan, Poland, Collegium Pharmaceuticum, Poznan, Poland
- Research and Implementation Unit, Greater Poland Cancer Center, Poznan, Poland
| | - Marcin Michalak
- Surgical, Oncological and Endoscopic Gynaecology Department, Greater Poland Cancer Center, Poznan, Poland
| | - Tomasz Kolenda
- Research and Implementation Unit, Greater Poland Cancer Center, Poznan, Poland
- Laboratory of Cancer Genetics, Greater Poland Cancer Center, Poznan, Poland
| | - Joanna Kozłowska-Masłoń
- Research and Implementation Unit, Greater Poland Cancer Center, Poznan, Poland
- Laboratory of Cancer Genetics, Greater Poland Cancer Center, Poznan, Poland
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Kacper Guglas
- Research and Implementation Unit, Greater Poland Cancer Center, Poznan, Poland
- Laboratory of Cancer Genetics, Greater Poland Cancer Center, Poznan, Poland
- Institute of Human Biology and Evolution, Faculty of Biology, Adam Mickiewicz University, Poznan, Poland
| | - Beata Stanisz
- Chair and Department of Pharmaceutical Chemistry, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
11
|
Hijazi MA, Gessner A, El-Najjar N. Repurposing of Chronically Used Drugs in Cancer Therapy: A Chance to Grasp. Cancers (Basel) 2023; 15:3199. [PMID: 37370809 DOI: 10.3390/cancers15123199] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/06/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Despite the advancement in drug discovery for cancer therapy, drug repurposing remains an exceptional opportunistic strategy. This approach offers many advantages (faster, safer, and cheaper drugs) typically needed to overcome increased challenges, i.e., side effects, resistance, and costs associated with cancer therapy. However, not all drug classes suit a patient's condition or long-time use. For that, repurposing chronically used medications is more appealing. This review highlights the importance of repurposing anti-diabetic and anti-hypertensive drugs in the global fight against human malignancies. Extensive searches of all available evidence (up to 30 March 2023) on the anti-cancer activities of anti-diabetic and anti-hypertensive agents are obtained from multiple resources (PubMed, Google Scholar, ClinicalTrials.gov, Drug Bank database, ReDo database, and the National Institutes of Health). Interestingly, more than 92 clinical trials are evaluating the anti-cancer activity of 14 anti-diabetic and anti-hypertensive drugs against more than 15 cancer types. Moreover, some of these agents have reached Phase IV evaluations, suggesting promising official release as anti-cancer medications. This comprehensive review provides current updates on different anti-diabetic and anti-hypertensive classes possessing anti-cancer activities with the available evidence about their mechanism(s) and stage of development and evaluation. Hence, it serves researchers and clinicians interested in anti-cancer drug discovery and cancer management.
Collapse
Affiliation(s)
- Mohamad Ali Hijazi
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Beirut Arab University, Beirut P.O. Box 11-5020, Lebanon
| | - André Gessner
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Nahed El-Najjar
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, 93053 Regensburg, Germany
| |
Collapse
|
12
|
Chang YL, Chou CH, Li YF, Huang LC, Kao Y, Hueng DY, Tsai CK. Antiproliferative and apoptotic effects of telmisartan in human glioma cells. Cancer Cell Int 2023; 23:111. [PMID: 37291545 DOI: 10.1186/s12935-023-02963-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 06/05/2023] [Indexed: 06/10/2023] Open
Abstract
Glioblastoma is the most common primary central nervous system tumor in adults. Angiotensin II receptor blockers (ARBs) are broadly applied to treat hypertension. Moreover, research has revealed that ARBs have the capacity to suppress the growth of several cancer types. In this study, we assessed the effects of three ARBs with the ability to cross the blood brain barrier (telmisartan, valsartan and fimasartan) on cell proliferation in three glioblastoma multiforme (GBM) cell lines. Telmisartan markedly suppressed the proliferation, migration, and invasion of these three GBM cell lines. Microarray data analysis revealed that telmisartan regulates DNA replication, mismatch repair, and the cell cycle pathway in GBM cells. Furthermore, telmisartan induced G0/G1 phase arrest and apoptosis. The bioinformatic analysis and western blotting results provide evidence that SOX9 is a downstream target of telmisartan. Telmisartan also suppressed tumor growth in vivo in an orthotopic transplant mouse model. Therefore, telmisartan is a potential treatment for human GBM.
Collapse
Affiliation(s)
- Yung-Lung Chang
- Department of Biochemistry, National Defense Medical Center, Taipei, 11490, Taiwan
| | - Chung-Hsing Chou
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, No. 325, Sec. 2, Cheng-Gong Road, Taipei, 11490, Taiwan
| | - Yao-Feng Li
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, 11490, Taiwan
| | - Li-Chun Huang
- Department of Biochemistry, National Defense Medical Center, Taipei, 11490, Taiwan
| | - Ying Kao
- Division of Neurosurgery, Department of Surgery, Taipei City Hospital Zhongxing Branch, Taipei, Taiwan
| | - Dueng-Yuan Hueng
- Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, 11490, Taiwan
| | - Chia-Kuang Tsai
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, No. 325, Sec. 2, Cheng-Gong Road, Taipei, 11490, Taiwan.
| |
Collapse
|
13
|
Antitumor Therapy Targeting the Tumor Microenvironment. JOURNAL OF ONCOLOGY 2023; 2023:6886135. [PMID: 36908706 PMCID: PMC10005879 DOI: 10.1155/2023/6886135] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 02/13/2023] [Accepted: 02/20/2023] [Indexed: 03/06/2023]
Abstract
The development and progression of tumors in human tissues extensively rely on its surrounding environment, that is, tumor microenvironment which includes a variety of cells, molecules, and blood vessels. These components are modified, organized, and integrated to support and facilitate the growth, invasion, and metabolism of tumor cells, suggesting them as potential therapeutic targets in anticancer treatment. An increasing number of pharmacological agents have been developed and clinically applied to target the oncogenic components in the tumor microenvironment, and in this review, we will summarize these pharmacological agents that directly or indirectly target the cellular or molecular components in the tumor microenvironment. However, difficulties and challenges still exist in this field, which will also be reported in this literature.
Collapse
|
14
|
Castro DV, Malhotra J, Meza L, Govindarajan A, Philip EJ, Pal SK. How to Treat Renal Cell Carcinoma. JACC CardioOncol 2022; 4:271-275. [PMID: 35818545 PMCID: PMC9270613 DOI: 10.1016/j.jaccao.2022.04.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 04/12/2022] [Accepted: 04/18/2022] [Indexed: 12/02/2022] Open
Abstract
With several newly approved regimens for RCC, cardiovascular risks must be considered during treatment selection. Although uncommon, toxicities associated with these systemic therapies can have shortterm and long-term implications. Although some toxicities may be lethal, others can be managed with early intervention. As further agents are developed, multidisciplinary expertise is needed to manage associated cardiovascular toxicities.
Collapse
Affiliation(s)
| | - Jasnoor Malhotra
- City of Hope Comprehensive Cancer Center, Duarte, California, USA
| | - Luis Meza
- City of Hope Comprehensive Cancer Center, Duarte, California, USA
| | | | - Errol J. Philip
- University of California-San Francisco, San Francisco, California, USA
| | - Sumanta K. Pal
- City of Hope Comprehensive Cancer Center, Duarte, California, USA
- Address for correspondence: Dr Sumanta K. Pal, City of Hope Comprehensive Cancer Center, 1500 East Duarte Road, Duarte, California 91010, USA. @montypal
| |
Collapse
|
15
|
Drobni ZD, Michielin O, Quinaglia T, Zlotoff DA, Zubiri L, Gilman HK, Supraja S, Merkely B, Muller V, Sullivan RJ, Reynolds KL, Pittet MJ, Jain RK, Neilan TG. Renin-angiotensin-aldosterone system inhibitors and survival in patients with hypertension treated with immune checkpoint inhibitors. Eur J Cancer 2022; 163:108-118. [PMID: 35065368 PMCID: PMC9618285 DOI: 10.1016/j.ejca.2021.12.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 11/17/2021] [Accepted: 12/21/2021] [Indexed: 01/06/2023]
Abstract
BACKGROUND Preclinical studies indicate that the concurrent use of inhibitors of the renin-angiotensin-aldosterone system (RAAS) may improve outcomes in broad groups of patients with cancer. There are limited data on the association between the use of RAAS inhibitors and outcomes among patients treated with immune checkpoint inhibitors (ICIs). METHODS We performed a retrospective study of all patients treated with an ICI in a single academic network. Of 10,903 patients, 5910 were on any anti-hypertensive medication. Of those on anti-hypertensive therapy, 3426 were prescribed a RAAS inhibitor during ICI treatment, and 2484 were prescribed other anti-hypertensive medications. The primary outcome was overall survival in the entire cohort and in sub-groups by cancer types. RESULTS Thoracic cancer (34%) and melanoma (16%) were the most common types of cancer. Those prescribed a RAAS inhibitor were older, more frequently male, and had more cardiovascular risk factors. In a Cox proportional hazard model, the concurrent use of RAAS inhibitors was associated with better overall survival (hazard ratio (HR):0.92, [95% Confidence Interval (CI):0.85-0.99], P = .032). Patients with gastrointestinal (HR:0.82, [95% CI: 0.67-1.01], P = .057) and genitourinary cancer (HR:0.81, [95% CI:0.64-1.01], P = .067) had a non-statistically significant better overall survival. CONCLUSIONS In this large retrospective study, patients with hypertension who were concomitantly taking a RAAS inhibitor during ICI therapy had better overall survival. This benefit was primarily noted among patients with gastrointestinal and genitourinary cancers. Prospective randomized trials are warranted to further evaluate and specify the benefit of RAAS inhibitors in patients with cancer who receive ICI therapy.
Collapse
Affiliation(s)
- Zsofia D. Drobni
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary,Cardiovascular Imaging Research Center (CIRC), Department of Radiology and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA,Corresponding author: Heart and Vascular Center, Semmelweis University, Városmajor street 68, Budapest, 1122, Hungary. , (Z.D. Drobni). @zsofidrobni (Z.D. Drobni)
| | - Olivier Michielin
- Oncology Department, Precision Oncology Center, Lausanne, Switzerland,Oncology Department, Lausanne University Hospital, Lausanne, Switzerland
| | - Thiago Quinaglia
- Cardiovascular Imaging Research Center (CIRC), Department of Radiology and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Daniel A. Zlotoff
- Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Leyre Zubiri
- Division of Oncology and Hematology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hannah K. Gilman
- Cardiovascular Imaging Research Center (CIRC), Department of Radiology and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sama Supraja
- Cardiovascular Imaging Research Center (CIRC), Department of Radiology and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Bela Merkely
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Veronika Muller
- Department of Pulmonology, Semmelweis University, Budapest, Hungary
| | - Ryan J. Sullivan
- Division of Oncology and Hematology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Kerry L. Reynolds
- Division of Oncology and Hematology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael J. Pittet
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Rakesh K. Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Tomas G. Neilan
- Cardiovascular Imaging Research Center (CIRC), Department of Radiology and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA,Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
16
|
Fiala O, Ostašov P, Rozsypalová A, Hora M, Šorejs O, Šustr J, Bendová B, Trávníček I, Filipovský J, Fínek J, Büchler T. Impact of Concomitant Cardiovascular Medication on Survival of Metastatic Renal Cell Carcinoma Patients Treated with Sunitinib or Pazopanib in the First Line. Target Oncol 2021; 16:643-652. [PMID: 34363554 DOI: 10.1007/s11523-021-00829-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/09/2021] [Indexed: 12/18/2022]
Abstract
BACKGROUND Patients with metastatic renal cell carcinoma (mRCC) are often elderly and have various comorbidities, including cardiovascular diseases. Although these patients have extensive co-exposure to targeted therapy and cardiovascular drugs, the impact of this co-exposure on outcomes for patients with mRCC remains unclear. OBJECTIVE Our objective was to evaluate the association between the use of cardiovascular medication and survival of patients with mRCC. METHODS The study included 343 consecutive patients with mRCC treated with sunitinib or pazopanib in the first line. Clinical data obtained from the Renal Cell Carcinoma Information System (RENIS) clinical registry and hospital information systems were retrospectively analyzed. Progression-free survival (PFS) and overall survival (OS) were compared according to the use of common medications, including antihypertensives (i.e., β-blockers [BBs], angiotensin-converting enzyme inhibitors, angiotensin II receptor blockers, calcium channel blockers, and diuretics), acetylsalicylic acid (aspirin), statins, and proton pump inhibitors. RESULTS The univariate Cox analysis evaluating the impact of the assessed comedications on patient survival revealed that only BBs were significantly associated with PFS (hazard ratio [HR] 0.533, p < 0.001) and OS (HR 0.641, p = 0.006). The median PFS and OS for users of BBs was 18.39 and 37.60 months versus 8.16 and 20.4 months for patients not using BBs (p < 0.001 and p < 0.001, respectively). The Cox multivariate analysis showed that the use of BBs was a significant factor for both PFS (HR 0.428, p = 0.001) and OS (HR 0.518, p = 0.001). CONCLUSIONS The results of this retrospective study suggest that the use of BBs is associated with favorable outcomes for patients with mRCC treated with sunitinib or pazopanib in the first line.
Collapse
Affiliation(s)
- Ondřej Fiala
- Department of Oncology and Radiotherapy, Faculty of Medicine and University Hospital in Pilsen, Charles University, alej Svobody 80, 304 60, Pilsen, Czech Republic.
- Laboratory of Cancer Treatment and Tissue Regeneration, Faculty of Medicine in Pilsen, Biomedical Center, Charles University, alej Svobody 76, Pilsen, Czech Republic.
| | - Pavel Ostašov
- Laboratory of Tumor Biology and Immunotherapy, Faculty of Medicine in Pilsen, Biomedical Center, Charles University, alej Svobody 76, Pilsen, Czech Republic
| | - Aneta Rozsypalová
- Department of Oncology, First Faculty of Medicine, Charles University and Thomayer Hospital, Videnska 800, Prague, Czech Republic
| | - Milan Hora
- Department of Urology, Faculty of Medicine and University Hospital in Pilsen, Charles University, E. Beneše 13, Pilsen, Czech Republic
| | - Ondřej Šorejs
- Department of Oncology and Radiotherapy, Faculty of Medicine and University Hospital in Pilsen, Charles University, alej Svobody 80, 304 60, Pilsen, Czech Republic
- Laboratory of Cancer Treatment and Tissue Regeneration, Faculty of Medicine in Pilsen, Biomedical Center, Charles University, alej Svobody 76, Pilsen, Czech Republic
| | - Jan Šustr
- Department of Oncology and Radiotherapy, Faculty of Medicine and University Hospital in Pilsen, Charles University, alej Svobody 80, 304 60, Pilsen, Czech Republic
| | - Barbora Bendová
- Department of Urology, Faculty of Medicine and University Hospital in Pilsen, Charles University, E. Beneše 13, Pilsen, Czech Republic
| | - Ivan Trávníček
- Department of Urology, Faculty of Medicine and University Hospital in Pilsen, Charles University, E. Beneše 13, Pilsen, Czech Republic
| | - Jan Filipovský
- 2nd Department of Internal Medicine, Faculty of Medicine and University Hospital in Pilsen, Charles University, E. Beneše 13, Pilsen, Czech Republic
| | - Jindřich Fínek
- Department of Oncology and Radiotherapy, Faculty of Medicine and University Hospital in Pilsen, Charles University, alej Svobody 80, 304 60, Pilsen, Czech Republic
| | - Tomáš Büchler
- Department of Oncology, First Faculty of Medicine, Charles University and Thomayer Hospital, Videnska 800, Prague, Czech Republic
| |
Collapse
|
17
|
Janisch F, Klotzbücher T, Marks P, Kienapfel C, Meyer CP, Yu H, Fühner C, Hillemacher T, Mori K, Mostafei H, Shariat SF, Fisch M, Dahlem R, Rink M. Predictive value of De Ritis ratio in metastatic renal cell carcinoma treated with tyrosine-kinase inhibitors. World J Urol 2021; 39:2977-2985. [PMID: 33649869 PMCID: PMC8405478 DOI: 10.1007/s00345-021-03628-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 02/03/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Predictive markers can help tailor treatment to the individual in metastatic renal cell carcinoma (mRCC). De Ritis ratio (DRR) is associated with oncologic outcomes in various solid tumors. OBJECTIVE To assess the value of DRR in prognosticating survival in mRCC patients treated with tyrosine-kinase inhibitors (TKI). METHODS Overall, 220 mRCC patients treated with TKI first-line therapy were analyzed. An optimal cut-off point for DRR was determined with Youden's J. We used multiple strata for DRR, performed descriptive, Kaplan-Meier and multivariable Cox-regression analyses to assess associations of DRR with progression-free (PFS) and overall survival (OS). RESULTS Patients above the optimal cut-off point for DRR of ≥ 1.58 had fewer liver metastases (p = 0.01). There was no difference in PFS (p > 0.05) between DRR groups. DRR above the median of 1.08 (HR 1.42; p = 0.03), DRR ≥ 1.1(HR 1.44; p = 0.02), ≥ 1.8 (HR 1.56; p = 0.03), ≥ 1.9 (HR 1.59; p = 0.02) and ≥ 2.0 (HR 1.63; p = 0.047) were associated with worse OS. These associations did not remain after multivariable adjustment. In the intermediate MSKCC group, DRR was associated with inferior OS at cut-offs ≥ 1.0 (HR 1.78; p = 0.02), ≥ 1.1 (HR 1.81; p = 0.01) and above median (HR 1.88; p = 0.007) in multivariable analyses. In patients with clear-cell histology, DRR above median (HR 1.54; p = 0.029) and DRR ≥ 1.1 (HR 1.53; p = 0.029) were associated with OS in multivariable analyses. CONCLUSION There was no independent association between DRR and survival of mRCC patients treated with TKI in the entire cohort. However, OS of patients with intermediate risk and clear-cell histology were affected by DRR. DRR could be used for tailored decision-making in these subgroups.
Collapse
Affiliation(s)
- Florian Janisch
- Department of Urology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
- Department of Urology, Medical University of Vienna, Vienna, Austria
| | - Thomas Klotzbücher
- Department of Urology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Phillip Marks
- Department of Urology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Christina Kienapfel
- Department of Urology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Christian P Meyer
- Department of Urology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Hang Yu
- Department of Urology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Constantin Fühner
- Department of Urology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Tobias Hillemacher
- Department of Urology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Keiichiro Mori
- Department of Urology, Medical University of Vienna, Vienna, Austria
- Department of Urology, Jikei University School of Medicine, Tokyo, Japan
| | - Hadi Mostafei
- Department of Urology, Medical University of Vienna, Vienna, Austria
- Department of Urology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shahrokh F Shariat
- Department of Urology, Medical University of Vienna, Vienna, Austria
- Institute for Urology and Reproductive Health, Sechenov University, Moscow, Russia
- Department of Urology, Weill Cornell Medical School, New York, NY, USA
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Karl Landsteiner Institute of Urology and Andrology, Vienna, Austria
- Department of Urology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Margit Fisch
- Department of Urology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Roland Dahlem
- Department of Urology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Michael Rink
- Department of Urology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.
| |
Collapse
|
18
|
Khanna P, Soh HJ, Chen CH, Saxena R, Amin S, Naughton M, Joslin PN, Moore A, Bakouny Z, O'Callaghan C, Catalano P, Signoretti S, McKay R, Choueiri TK, Bhasin M, Walther T, Bhatt RS. ACE2 abrogates tumor resistance to VEGFR inhibitors suggesting angiotensin-(1-7) as a therapy for clear cell renal cell carcinoma. Sci Transl Med 2021; 13:13/577/eabc0170. [PMID: 33472951 DOI: 10.1126/scitranslmed.abc0170] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 09/24/2020] [Indexed: 12/24/2022]
Abstract
Angiotensin converting enzyme 2 (ACE2) is an enzyme that belongs to the renin-angiotensin system (RAS) and antagonizes the classical angiotensin (Ang) II/angiotensin II receptor type 1 (AT1) receptor pathway. Here, we report that higher ACE2 expression correlates with better overall survival in patients with clear cell renal cell carcinoma (ccRCC). Moreover, ACE2 has inhibitory effects on tumor proliferation in ccRCC in vitro and in preclinical animal models of ccRCC. We further show that Ang-(1-7), a heptapeptide generated by ACE2, is the likely mediator of this effect. Vascular endothelial growth factor receptor-tyrosine kinase inhibitor (VEGFR-TKI) treatment of ccRCC xenografts decreased ACE2 expression, and combination treatment with VEGFR-TKI and Ang-(1-7) generated additive suppression of tumor growth and improved survival outcomes. Last, the addition of Ang-(1-7) to programmed death-ligand 1 (PD-L1) pathway inhibitor and VEGFR-TKI showed further growth suppression in an immunocompetent RCC model. Together, these results suggest that targeting the ACE2/Ang-(1-7) axis is a promising therapeutic strategy against ccRCC.
Collapse
Affiliation(s)
- Prateek Khanna
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Department of Medicine, Mount Auburn Hospital, Harvard Medical School, Cambridge, MA 02138, USA
| | - Hong Jie Soh
- Department of Pharmacology and Therapeutics, School of Medicine and School of Pharmacy, University College Cork, Cork T12 K8AF, Ireland
| | - Chun-Hau Chen
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Ruchi Saxena
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Seema Amin
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Maura Naughton
- Department of Pharmacology and Therapeutics, School of Medicine and School of Pharmacy, University College Cork, Cork T12 K8AF, Ireland
| | - Patrick Neset Joslin
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Andrew Moore
- Department of Pharmacology and Therapeutics, School of Medicine and School of Pharmacy, University College Cork, Cork T12 K8AF, Ireland
| | - Ziad Bakouny
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Carol O'Callaghan
- Department of Pharmacology and Therapeutics, School of Medicine and School of Pharmacy, University College Cork, Cork T12 K8AF, Ireland
| | - Paul Catalano
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Sabina Signoretti
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.,Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Rana McKay
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Toni K Choueiri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Manoj Bhasin
- Division of Interdisciplinary Medicine and Biotechnology and Genomics, Proteomics, Bioinformatics and Systems Biology Center, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Thomas Walther
- Department of Pharmacology and Therapeutics, School of Medicine and School of Pharmacy, University College Cork, Cork T12 K8AF, Ireland. .,Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, Greifswald 17489, Germany
| | - Rupal S Bhatt
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
19
|
Neophytou CM, Panagi M, Stylianopoulos T, Papageorgis P. The Role of Tumor Microenvironment in Cancer Metastasis: Molecular Mechanisms and Therapeutic Opportunities. Cancers (Basel) 2021; 13:cancers13092053. [PMID: 33922795 PMCID: PMC8122975 DOI: 10.3390/cancers13092053] [Citation(s) in RCA: 165] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Metastasis, the process by which cancer cells escape primary tumor site and colonize distant organs, is responsible for most cancer-related deaths. The tumor microenvironment (TME), comprises different cell types, including immune cells and cancer-associated fibroblasts, as well as structural elements, such as collagen and hyaluronan that constitute the extracellular matrix (ECM). Intratumoral interactions between the cellular and structural components of the TME regulate the aggressiveness, and dissemination of malignant cells and promote immune evasion. At the secondary site, the TME also facilitates escape from dormancy to enhance metastatic tumor outgrowth. Moreover, the ECM applies mechanical forces on tumors that contribute to hypoxia and cancer cell invasiveness whereas also hinders drug delivery and efficacy in both primary and metastatic sites. In this review, we summarize the latest developments regarding the role of the TME in cancer progression and discuss ongoing efforts to remodel the TME to stop metastasis in its tracks. Abstract The tumor microenvironment (TME) regulates essential tumor survival and promotion functions. Interactions between the cellular and structural components of the TME allow cancer cells to become invasive and disseminate from the primary site to distant locations, through a complex and multistep metastatic cascade. Tumor-associated M2-type macrophages have growth-promoting and immunosuppressive functions; mesenchymal cells mass produce exosomes that increase the migratory ability of cancer cells; cancer associated fibroblasts (CAFs) reorganize the surrounding matrix creating migration-guiding tracks for cancer cells. In addition, the tumor extracellular matrix (ECM) exerts determinant roles in disease progression and cancer cell migration and regulates therapeutic responses. The hypoxic conditions generated at the primary tumor force cancer cells to genetically and/or epigenetically adapt in order to survive and metastasize. In the circulation, cancer cells encounter platelets, immune cells, and cytokines in the blood microenvironment that facilitate their survival and transit. This review discusses the roles of different cellular and structural tumor components in regulating the metastatic process, targeting approaches using small molecule inhibitors, nanoparticles, manipulated exosomes, and miRNAs to inhibit tumor invasion as well as current and future strategies to remodel the TME and enhance treatment efficacy to block the detrimental process of metastasis.
Collapse
Affiliation(s)
- Christiana M. Neophytou
- European University Research Center, Nicosia 2404, Cyprus;
- Tumor Microenvironment, Metastasis and Experimental Therapeutics Laboratory, Basic and Translational Cancer Research Center, Department of Life Sciences, European University Cyprus, Nicosia 1516, Cyprus
| | - Myrofora Panagi
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia 2109, Cyprus; (M.P.); (T.S.)
| | - Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia 2109, Cyprus; (M.P.); (T.S.)
| | - Panagiotis Papageorgis
- European University Research Center, Nicosia 2404, Cyprus;
- Tumor Microenvironment, Metastasis and Experimental Therapeutics Laboratory, Basic and Translational Cancer Research Center, Department of Life Sciences, European University Cyprus, Nicosia 1516, Cyprus
- Correspondence: ; Tel.: +357-22-713158
| |
Collapse
|
20
|
Siljee S, Milne B, Brasch HD, Bockett N, Patel J, Davis PF, Kennedy-Smith A, Itinteang T, Tan ST. Expression of Components of the Renin-Angiotensin System by Cancer Stem Cells in Renal Clear Cell Carcinoma. Biomolecules 2021; 11:537. [PMID: 33916968 PMCID: PMC8067590 DOI: 10.3390/biom11040537] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/31/2021] [Accepted: 04/03/2021] [Indexed: 12/23/2022] Open
Abstract
This study investigated the expression of components of the renin-angiotensin system (RAS) by cancer stem cells (CSCs) we have recently demonstrated in renal clear cell carcinoma (RCCC). Fifteen RCCC tissue samples underwent immunohistochemical staining for components of the RAS: renin, pro-renin receptor (PRR), angiotensin-converting enzyme (ACE), angiotensin-converting enzyme 2 (ACE2), and angiotensin II receptor 2 (AT2R). Immunofluorescence co-staining or double immunohistochemical staining of these components of the RAS with stemness-associated markers OCT4 or KLF4 was performed on two of the samples. Protein and transcript expression of these components of the RAS in six RCCC tissue samples was investigated using western blotting and reverse transcription quantitative polymerase chain reaction (RT-qPCR), respectively. In addition, angiotensin II receptor 1 (AT1R) was investigated using RT-qPCR only. Immunohistochemical staining demonstrated expression of renin, PRR, and ACE2 in 11, 13, and 13 out of 15 RCCC samples, respectively, while AT2R was expressed in all 15 samples. ACE was detected in the endothelium of normal vasculature only. Double immunohistochemical staining demonstrated localization of ACE2, but not renin, to the KLF4+ CSCs. Immunofluorescence staining showed localization of PRR and AT2R to the OCT4+ CSCs. Western blotting confirmed protein expression of all components of the RAS except renin. RT-qPCR demonstrated transcript expression of all components of the RAS including AT1R, but not AT2R, in all six RCCC tissue samples. This study demonstrated expression of PRR, ACE2, and AT2R by the CSCs within RCCC. Further studies may lead to novel therapeutic targeting of CSCs by manipulation of the RAS in the treatment of this aggressive cancer.
Collapse
Affiliation(s)
- Sam Siljee
- Gillies McIndoe Research Institute, Wellington 6242, New Zealand; (S.S.); (B.M.); (H.D.B.); (N.B.); (J.P.); (P.F.D.); (T.I.)
| | - Bridget Milne
- Gillies McIndoe Research Institute, Wellington 6242, New Zealand; (S.S.); (B.M.); (H.D.B.); (N.B.); (J.P.); (P.F.D.); (T.I.)
| | - Helen D. Brasch
- Gillies McIndoe Research Institute, Wellington 6242, New Zealand; (S.S.); (B.M.); (H.D.B.); (N.B.); (J.P.); (P.F.D.); (T.I.)
| | - Nicholas Bockett
- Gillies McIndoe Research Institute, Wellington 6242, New Zealand; (S.S.); (B.M.); (H.D.B.); (N.B.); (J.P.); (P.F.D.); (T.I.)
| | - Josie Patel
- Gillies McIndoe Research Institute, Wellington 6242, New Zealand; (S.S.); (B.M.); (H.D.B.); (N.B.); (J.P.); (P.F.D.); (T.I.)
| | - Paul F. Davis
- Gillies McIndoe Research Institute, Wellington 6242, New Zealand; (S.S.); (B.M.); (H.D.B.); (N.B.); (J.P.); (P.F.D.); (T.I.)
| | - Andrew Kennedy-Smith
- Department of Urology, Wellington Regional Hospital, Wellington 6021, New Zealand;
| | - Tinte Itinteang
- Gillies McIndoe Research Institute, Wellington 6242, New Zealand; (S.S.); (B.M.); (H.D.B.); (N.B.); (J.P.); (P.F.D.); (T.I.)
| | - Swee T. Tan
- Gillies McIndoe Research Institute, Wellington 6242, New Zealand; (S.S.); (B.M.); (H.D.B.); (N.B.); (J.P.); (P.F.D.); (T.I.)
- Wellington Regional Plastic, Maxillofacial and Burns Unit, Hutt Hospital, Lower Hutt 5010, New Zealand
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC 3010, Australia
| |
Collapse
|
21
|
Liu F, Li L, Chen J, Wu Y, Cao Y, Zhong P. A Network Pharmacology to Explore the Mechanism of Calculus Bovis in the Treatment of Ischemic Stroke. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6611018. [PMID: 33778069 PMCID: PMC7972848 DOI: 10.1155/2021/6611018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/15/2021] [Accepted: 02/20/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Calculus Bovis is a valuable Chinese medicine, which is widely used in the clinical treatment of ischemic stroke. The present study is aimed at investigating its target and the mechanism involved in ischemic stroke treatment by network pharmacology. METHODS Effective compounds of Calculus Bovis were collected using methods of network pharmacology and using the Bioinformatics Analysis Tool for Molecular Mechanism of Traditional Chinese Medicine (BATMAN-TCM) and the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP). Potential compound targets were searched in the TCMSP and SwissTargetPrediction databases. Ischemic stroke-related disease targets were searched in the Drugbank, DisGeNet, OMIM, and TTD databases. These two types of targets were uploaded to the STRING database, and a network of their interaction (PPI) was built with its characteristics calculated, aiming to reveal a number of key targets. Hub genes were selected using a plug-in of the Cytoscape software, and Gene Ontology (GO) biological processes and pathway enrichment analyses of Kyoto Encyclopedia of Genes and Genomes (KEGG) were conducted using the clusterProfiler package of R language. RESULTS Among 12 compounds, deoxycorticosterone, methyl cholate, and biliverdin were potentially effective components. A total of 344 Calculus Bovis compound targets and 590 ischemic stroke targets were found with 92 overlapping targets, including hub genes such as TP53, AKT, PIK2CA, MAPK3, MMP9, and MMP2. Biological functions of Calculus Bovis are associated with protein hydrolyzation, phosphorylation of serine/threonine residues of protein substrates, peptide bond hydrolyzation of peptides and proteins, hydrolyzation of intracellular second messengers, antioxidation and reduction, RNA transcription, and other biological processes. CONCLUSION Calculus Bovis may play a role in ischemic stroke by activating PI3K-AKT and MAPK signaling pathways, which are involved in regulating inflammatory response, cell apoptosis, and proliferation.
Collapse
Affiliation(s)
- Fangchen Liu
- Department of Neurology, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
- Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ling Li
- Institute of Vascular Disease, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Jian Chen
- Institute of Vascular Disease, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Ying Wu
- Department of Neurology, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Yongbing Cao
- Institute of Vascular Disease, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Ping Zhong
- Department of Neurology, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
- Department of Neurology, Shidong Hospital of Yangpu District, Shanghai 200090, China
| |
Collapse
|
22
|
Yang J, Yang X, Gao L, Zhang J, Yi C, Huang Y. The role of the renin-angiotensin system inhibitors in malignancy: a review. Am J Cancer Res 2021; 11:884-897. [PMID: 33791161 PMCID: PMC7994166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 01/12/2021] [Indexed: 06/12/2023] Open
Abstract
Hypertension is one of the most prevalent diseases in cardiology. The angiotensin receptor blockers (ARBs)/angiotensin converting enzyme inhibitors (ACEIs) are widely used drugs to stabilize the blood pressure via inhibition of the renin-angiotensin system (RAS). Studies have found that the exposure to RAS inhibitors (RASi) can suppress the development of cancers via multimodal mechanisms and has attracted increased attentions in the recent past. Owing the potential of RASi to inhibit tumor growth, proliferation and metastasis, they are considered as the potential and exciting candidates to enhance the effect of chemo-radiotherapy and targeted therapy efficacy. However, there are conflicting reports as to the use of ARB/ACEI in all facets of tumor growth. In this study, we comprehensively summarize and review the potential mechanisms of RASi in cancer treatment, like inhibition of tumor angiogenesis, reduction of cancer-associated fibroblasts (CAFs) and extracellular matrix (ECM), regulation of immune cells and improvement of hypoxia. Additionally, based on the basic and clinical experiments, we analyze the views and results regarding the role of RASi plays in tumor from genesis to recurrence, and certainly cancer treatment (chemo-radiotherapy and targeted therapy). In the last, not only do we discuss the prospects of using RASi to enhance cancer treatment efficacy but also point out the conflicting situation with the intention to give some directions and inspiration on this topic.
Collapse
Affiliation(s)
- Ju Yang
- West China School of Basic Medical Science and Forensic Medicine, Sichuan UniversityChengdu 610041, China
| | - Xi Yang
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan UniversityChengdu 610041, China
| | - Ling Gao
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan UniversityChengdu 610041, China
| | - Jie Zhang
- Lab of Experimental Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation CenterChengdu 610041, China
| | - Cheng Yi
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan UniversityChengdu 610041, China
| | - Ying Huang
- West China School of Basic Medical Science and Forensic Medicine, Sichuan UniversityChengdu 610041, China
| |
Collapse
|
23
|
Solano-Iturri JD, Echevarría E, Unda M, Loizaga-Iriarte A, Pérez-Fernández A, Angulo JC, López JI, Larrinaga G. Clinical Implications of (Pro)renin Receptor (PRR) Expression in Renal Tumours. Diagnostics (Basel) 2021; 11:diagnostics11020272. [PMID: 33578778 PMCID: PMC7916453 DOI: 10.3390/diagnostics11020272] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/05/2021] [Accepted: 02/08/2021] [Indexed: 01/01/2023] Open
Abstract
(1) Background: Renal cancer is one of the most frequent malignancies in Western countries, with an unpredictable clinical outcome, partly due to its high heterogeneity and the scarcity of reliable biomarkers of tumour progression. (Pro)renin receptor (PRR) is a novel receptor of the renin–angiotensin system (RAS) that has been associated with the development and progression of some solid tumours by RAS-dependent and -independent mechanisms. (2) Methods: In this study, we analysed the immunohistochemical expression of PRR at the centre and border in a series of 83 clear-cell renal cell (CCRCCs), 19 papillary (PRCC) and 7 chromophobe (ChRCC) renal cell carcinomas, and the benign tumour renal oncocytoma (RO, n = 11). (3) Results: PRR is expressed in all the tumour subtypes, with higher mean staining intensity in ChRCCs and ROs. A high expression of PRR at the tumour centre and at the infiltrative front of CCRCC tissues is significantly associated with high grade, tumour diameter, local invasion and stage, and with high mortality risk by UCLA integrated staging system (UISS) scale. (4) Conclusions: These findings indicate that PRR is associated with the development and progression of renal tumours. Its potential as a novel biomarker for RCC diagnosis/prognosis and as a promising therapeutic target should be taken into account in the future.
Collapse
Affiliation(s)
- Jon Danel Solano-Iturri
- Department of Pathology, Donostia University Hospital, 20014 Donostia/San Sebastian, Spain;
- Department of Medical-Surgical Specialities, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
- Biocruces-Bizkaia Health Research Institute, 48903 Barakaldo, Spain;
| | - Enrique Echevarría
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain;
| | - Miguel Unda
- Department of Urology, Basurto University Hospital, University of the Basque Country (UPV/EHU), 48013 Bilbao, Spain; (M.U.); (A.L.-I.); (A.P.-F.)
| | - Ana Loizaga-Iriarte
- Department of Urology, Basurto University Hospital, University of the Basque Country (UPV/EHU), 48013 Bilbao, Spain; (M.U.); (A.L.-I.); (A.P.-F.)
| | - Amparo Pérez-Fernández
- Department of Urology, Basurto University Hospital, University of the Basque Country (UPV/EHU), 48013 Bilbao, Spain; (M.U.); (A.L.-I.); (A.P.-F.)
| | - Javier C. Angulo
- Clinical Department. Faculty of Medical Sciences. European University of Madrid, 28905 Getafe, Spain;
| | - José I. López
- Biocruces-Bizkaia Health Research Institute, 48903 Barakaldo, Spain;
- Department of Pathology, Cruces University Hospital, 48903 Barakaldo, Spain
| | - Gorka Larrinaga
- Biocruces-Bizkaia Health Research Institute, 48903 Barakaldo, Spain;
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain;
- Department of Nursing, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
- Correspondence:
| |
Collapse
|
24
|
Renin-Angiotensin System in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1277:105-114. [PMID: 33119868 DOI: 10.1007/978-3-030-50224-9_7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
For enhancing the antitumor effects of current immunotherapies including immune-checkpoint blockade, it is important to reverse cancer-induced immunosuppression. The renin-angiotensin system (RAS) controls systemic body fluid circulation; however, the presence of a local RAS in tumors has been reported. Furthermore, the local RAS in tumors influences various immune and interstitial cells and affects tumor immune response. RAS stimulation through the angiotensin II type 1 receptor has been reported to inhibit tumor immune response. Therefore, RAS inhibitors and combined treatment with immunotherapy are expected in the future. In this chapter, we provide a background on the RAS and describe the tumor environment with regard to the RAS and tumor immune response.
Collapse
|
25
|
Liu Y, Zhu C, Guo J, Chen Y, Meng C. The Neuroprotective Effect of Irisin in Ischemic Stroke. Front Aging Neurosci 2020; 12:588958. [PMID: 33414714 PMCID: PMC7782245 DOI: 10.3389/fnagi.2020.588958] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 12/01/2020] [Indexed: 12/14/2022] Open
Abstract
Irisin is a PGC-1α-dependent myokine that causes increased energy expenditure by driving the development of white adipose tissue into brown fat-like tissue. Exercise can improve irisin levels and lead to its release into the blood. In ischemic stroke, neurons are always sensitive to energy supply; after a series of pathophysiological processes, reactive oxygen species that are detrimental to cell survival via mitochondrial dysfunction are generated in large quantities. As a protein associated with exercise, irisin can alleviate brain injury in the pathogenesis of ischemic stroke. It is thought that irisin can upregulate the levels of brain-derived neurotrophic factor (BDNF), which protects nerve cells from injury during ischemic stroke. Furthermore, the release of irisin into the blood via exercise influences the mitochondrial dynamics crucial to maintaining the normal function of nerve cells. Consequently, we intended to summarize the known effects of irisin during ischemic stroke.
Collapse
Affiliation(s)
- Yaqiang Liu
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Chunhua Zhu
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jiahui Guo
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yonghong Chen
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Chaoyue Meng
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
26
|
Volovat SR, Volovat C, Miron I, Kanbay M, Goldsmith D, Lungulescu C, Badarau SC, Covic A. Oncogenic mechanisms in renal insufficiency. Clin Kidney J 2020; 14:507-515. [PMID: 33623673 PMCID: PMC7886561 DOI: 10.1093/ckj/sfaa122] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Accepted: 05/08/2020] [Indexed: 12/15/2022] Open
Abstract
The prevalence of both cancer and end-stage renal disease is increasing. In addition, medical advances have meant increased survival rates for both diseases. Many chemotherapeutics are renally excreted, and conversely, renal insufficiency promotes a pro-neoplastic state, including genitourinary and other cancers. Dialysis prolongs life while increasing cancer risk. Proposed oncogenic mechanisms include immune dysfunction, chronic inflammation, changes in gut microbiota and stimulation of the renin-angiotensin system. This review summarizes current concepts in the relationship between cancer and renal insufficiency.
Collapse
Affiliation(s)
- Simona Ruxandra Volovat
- Department of Medical Oncology, University of Medicine and Pharmacy 'Grigore T Popa', Iasi, Romania
| | - Constantin Volovat
- Department of Medical Oncology, University of Medicine and Pharmacy 'Grigore T Popa', Iasi, Romania
| | - Ingrith Miron
- Department of Medical Oncology, University of Medicine and Pharmacy 'Grigore T Popa', Iasi, Romania
| | - Mehmet Kanbay
- Department of Nephrology, Division of Nephrology, Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - David Goldsmith
- Department of Nephrology, St George's University Hospital, London, UK
| | - Cristian Lungulescu
- Department of Medical Oncology, University of Medicine and Pharmacy of Craiova, Craiova, Romania
| | - Silvia Corina Badarau
- Department of Medical Oncology, University of Medicine and Pharmacy 'Grigore T Popa', Iasi, Romania
| | - Adrian Covic
- Department of Medical Oncology, University of Medicine and Pharmacy 'Grigore T Popa', Iasi, Romania
| |
Collapse
|
27
|
Angiotensin Inhibition, TGF-β and EMT in Cancer. Cancers (Basel) 2020; 12:cancers12102785. [PMID: 32998363 PMCID: PMC7601465 DOI: 10.3390/cancers12102785] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/24/2020] [Accepted: 09/24/2020] [Indexed: 12/12/2022] Open
Abstract
Angiotensin inhibitors are standard drugs in cardiovascular and renal diseases that have antihypertensive and antifibrotic properties. These drugs also exert their antifibrotic effects in cancer by reducing collagen and hyaluronan deposition in the tumor stroma, thus enhancing drug delivery. Angiotensin II signaling interferes with the secretion of the cytokine TGF-β-a known driver of malignancy. TGF-β stimulates matrix production in cancer-associated fibroblasts, and thus drives desmoplasia. The effect of TGF-β on cancer cells itself is stage-dependent and changes during malignant progression from inhibitory to stimulatory. The intracellular signaling for the TGF-β family can be divided into an SMAD-dependent canonical pathway and an SMAD-independent noncanonical pathway. These capabilities have made TGF-β an interesting target for numerous drug developments. TGF-β is also an inducer of epithelial-mesenchymal transition (EMT). EMT is a highly complex spatiotemporal-limited process controlled by a plethora of factors. EMT is a hallmark of metastatic cancer, and with its reversal, an important step in the metastatic cascade is characterized by a loss of epithelial characteristics and/or the gain of mesenchymal traits.
Collapse
|
28
|
Abstract
BACKGROUND Cardio-oncology aims to mitigate adverse cardiovascular manifestations in cancer survivors, but treatment-induced hypertension or aggravated hypertension has received less attention in these high cardiovascular risk patients. METHODS In this systematic review, we searched literature for contemporary data on the prevalence, pathophysiologic mechanisms, treatment implications and preventive strategies of hypertension in patients under antineoplastic therapy. RESULTS Several classes of antineoplastic drugs, including mainly vascular endothelial growth factor inhibitors, proteasome inhibitors, cisplatin derivatives, corticosteroids or radiation therapy were consistently associated with increased odds for new-onset hypertension or labile hypertensive status in previous controlled patients. Moreover, hypertension constitutes a major risk factor for chemotherapy-induced cardiotoxicity, which is the most serious cardiovascular adverse effect of antineoplastic therapy. Despite the heterogeneity of pooled studies, the pro-hypertensive profile of examined drug classes could be attributed to common structural and functional disorders. Importantly, certain antihypertensive drugs are considered to be more effective in the management of hypertension in this population and may partially attenuate indirect complications of cancer treatment, such as progressive development of cardiomyopathy and/or cardiovascular death. Nonpharmacological approaches to alleviate hypertension in cancer patients are also described, albeit adjudicated as less effective in general. CONCLUSION A growing body of evidence suggests that multiple antineoplastic agents increase the rate of progression of hypertension. Physicians need to balance the life-saving cancer treatment and the inflated risk of adverse cardiovascular events due to suboptimal management of hypertension in order to achieve improved clinical outcomes and sustained survival for their patients.
Collapse
|
29
|
Hauge A, Rofstad EK. Antifibrotic therapy to normalize the tumor microenvironment. J Transl Med 2020; 18:207. [PMID: 32434573 PMCID: PMC7240990 DOI: 10.1186/s12967-020-02376-y] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 05/13/2020] [Indexed: 02/07/2023] Open
Abstract
Most tumors develop abnormal fibrotic regions consisting of fibroblasts, immune cells, and a dense extracellular matrix (ECM) immersed in a viscous interstitial fluid, and an abundant fibrotic tumor microenvironment (TME) is associated with poor outcome of treatment. It has been hypothesized that the treatment of cancer may be improved by interventions aiming to normalize this TME. The approaches used in attempts to normalize the fibrotic TME can be categorized into three strategies of targeted antifibrotic therapy: targeting of components of the ECM, targeting of the producers of the ECM components-the activated cancer-associated fibroblasts (CAFs), and targeting of the signaling pathways activating CAFs. To target the ECM, enzymes against components of the ECM have been used, including collagenase, relaxin, hyaluronidase, and lyxyl oxidase. Targeting of CAFs have been investigated by using agents aiming to eliminate or reprogram CAFs. CAFs are activated primarily by transforming growth factor-β (TGF-β), hedgehog, or focal adhesion kinase signaling, and several agents have been used to target these signaling pathways, including angiotensin II receptor I blockers (e.g., losartan) to inhibit the TGF-β pathway. Taken together, these studies have revealed that antifibrotic therapy is a two-edged sword: while some studies suggest enhanced response to treatment after antifibrotic therapy, others suggest that antifibrotic therapy may lead to increased tumor growth, metastasis, and impaired outcome of treatment. There are several possible explanations of these conflicting observations. Most importantly, tumors contain different subpopulations of CAFs, and while some subpopulations may promote tumor growth and metastasis, others may inhibit malignant progression. Furthermore, the outcome of antifibrotic therapy may depend on stage of disease, duration of treatment, treatment-induced activation of alternative profibrotic signaling pathways, and treatment-induced recruitment of tumor-supporting immune cells. Nevertheless, losartan-induced suppression of TGF-β signaling appears to be a particularly promising strategy. Losartan is a widely prescribed antihypertensive drug and highly advantageous therapeutic effects have been observed after losartan treatment of pancreatic cancer. However, improved understanding of the mechanisms governing the development of fibrosis in tumors is needed before safe antifibrotic treatments can be established.
Collapse
Affiliation(s)
- Anette Hauge
- Group of Radiation Biology and Tumor Physiology, Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Einar K Rofstad
- Group of Radiation Biology and Tumor Physiology, Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
30
|
Therapeutic potential of renin angiotensin system inhibitors in cancer cells metastasis. Pathol Res Pract 2020; 216:153010. [PMID: 32534713 DOI: 10.1016/j.prp.2020.153010] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 04/13/2020] [Accepted: 05/10/2020] [Indexed: 12/15/2022]
Abstract
Metastasis is a complex process which contributes to the dissemination of cancer cells to other organs and forms new tumor sites. The proliferation of tumor cells is a necessary step for the initiation and progression of cancers and is associated with the formation of new vessels. In the latter stages of metastasis, cancer cells may spread into the extracellular matrix and may form metastatic nodules. Despite efforts to prevent this, effective therapies are limited in the treatment of some malignancies. Among the different tumor properties which could be usefully employed as a cancer target, metastasis may be one suitable target. The renin- angiotensin system is a physiological pathway that contributes to the proliferation of tumor cells, angiogenesis and the inflammatory response in tumor tissue. Angiotensin II (ANGII), a key peptide of this pathway, induces cell proliferation through the activation of two cellular pathways (mitogen-activated protein kinase (MAPK)-STAT3 and phosphoinositide 3-kinase (PI3K) -AKT pathway). AT1-R increases angiogenesis via the elevation of angiogenic factors expression (vascular endothelial growth factor (VEGF) and matrix metallopeptidases (MMPs)). The local activation of the RAS pathway increases the expression of ICAM, VCAM and MMPs genes that are involved in the late steps of the metastasis process. There is some evidence that RAS components are expressed in metastatic tumors and RASIs (renin-angiotensin system inhibitors) could be used to reduce cancer metastasis by affecting the mechanisms involved in several different cancers. Therefore, we have summarized the effects of RASIs, observed in pre-clinical and clinical studies of cancer cell metastasis.
Collapse
|
31
|
D'alessandro M, Bergantini L, Carleo A, Cameli P, Perrone A, Fossi A, Sestini P, Bargagli E. Neutrophil-to-lymphocyte ratio in bronchoalveolar lavage from IPF patients: a novel prognostic biomarker? Minerva Med 2020; 113:526-531. [PMID: 32407050 DOI: 10.23736/s0026-4806.20.06614-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND This is the first time that a bronchoalveolar lavage (BAL) neutrophil-to-lymphocyte ratio (NL-ratio) has been demonstrated in sarcoidosis and chronic Hypersensitivity Pneumonitis (cHP) than in idiopathic pulmonary fibrosis (IPF) patients. SUBJECTS AND METHODS Consecutive BAL samples from the 167 interstitial lung disease (ILD) patients were retrospectively enrolled in the study and clustered into three diagnostic categories: IPF, cHP and sarcoidosis. RESULTS NL-ratio which proved higher in IPF (mean±SD, 2.1±3.8) than sarcoidosis (mean±SD 0.7±1.9; p<1E-04) and cHP patients (mean±SD 1.6±3.1; p=7.7E-03). ROC curve analysis to discriminate between Sarcoidosis and other ILDs showed an area under the curve (AUC) of 83.7%, (56% sensitivity and 96% specificity) while IPF and the other ILD were discriminated with AUC of 73% using a NL-ratio threshold value of 0.48 (73% sensitivity and 63% specificity). Interestingly, the NL-ratio was significantly correlated with other prognostic parameters: it was inversely correlated with forced vital capacity (FVC) (r=-0.3; p=2.5E-02) and forced expiration volume in 1 second (FEV1) (r=-0.3; p=2E-02) percentages and directly correlated with composite pulmonary index (CPI) score (r=0.3; p=3.2E-02). A decision-tree statistical algorithm was applied. CONCLUSIONS This is the first time that a lower NL-ratio has been demonstrated in sarcoidosis and cHP than in IPF patients. The present preliminary report indicates a relationship between BAL NL- ratio and lung function parameters in patients with IPF: this ratio may help to optimize management of IPF patients and to improve follow-up and outcome.
Collapse
Affiliation(s)
- Miriana D'alessandro
- Respiratory Diseases and Lung Transplantation, Department of Medical and Surgical Sciences & Neurosciences, Siena University Hospital, Siena, Italy -
| | - Laura Bergantini
- Respiratory Diseases and Lung Transplantation, Department of Medical and Surgical Sciences & Neurosciences, Siena University Hospital, Siena, Italy
| | - Alfonso Carleo
- Department of Pulmonology, Hannover Medical School, Hannover, Germany
| | - Paolo Cameli
- Respiratory Diseases and Lung Transplantation, Department of Medical and Surgical Sciences & Neurosciences, Siena University Hospital, Siena, Italy
| | - Anna Perrone
- Respiratory Diseases and Lung Transplantation, Department of Medical and Surgical Sciences & Neurosciences, Siena University Hospital, Siena, Italy
| | - Antonella Fossi
- Respiratory Diseases and Lung Transplantation, Department of Medical and Surgical Sciences & Neurosciences, Siena University Hospital, Siena, Italy
| | - Piersante Sestini
- Respiratory Diseases and Lung Transplantation, Department of Medical and Surgical Sciences & Neurosciences, Siena University Hospital, Siena, Italy
| | - Elena Bargagli
- Respiratory Diseases and Lung Transplantation, Department of Medical and Surgical Sciences & Neurosciences, Siena University Hospital, Siena, Italy
| |
Collapse
|
32
|
He Z, Ning N, Zhou Q, Khoshnam SE, Farzaneh M. Mitochondria as a therapeutic target for ischemic stroke. Free Radic Biol Med 2020; 146:45-58. [PMID: 31704373 DOI: 10.1016/j.freeradbiomed.2019.11.005] [Citation(s) in RCA: 146] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 10/07/2019] [Accepted: 11/03/2019] [Indexed: 12/24/2022]
Abstract
Stroke is the leading cause of death and physical disability worldwide. Mitochondrial dysfunction has been considered as one of the hallmarks of ischemic stroke and contributes to the pathology of ischemia and reperfusion. Mitochondria is essential in promoting neural survival and neurological improvement following ischemic stroke. Therefore, mitochondria represent an important drug target for stroke treatment. This review discusses the mitochondrial molecular mechanisms underlying cerebral ischemia and involved in reactive oxygen species generation, mitochondrial electron transport dysfunction, mitochondria-mediated regulation of inflammasome activation, mitochondrial dynamics and biogenesis, and apoptotic cell death. We highlight the potential of mitochondrial transfer by stem cells as a therapeutic target for stroke treatment and provide valuable insights for clinical strategies. A better understanding of the roles of mitochondria in ischemia-induced cell death and protection may provide a rationale design of novel therapeutic interventions in the ischemic stroke.
Collapse
Affiliation(s)
- Zhi He
- Department of Pharmacy, Luohe Medical College, Luohe, 462000, China
| | - Niya Ning
- Department of Obstetrics and Gynecology, Shaoling District People's Hospital of Luohe City, Luohe, 462300, China
| | - Qiongxiu Zhou
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Chengdu, 610052, China.
| | - Seyed Esmaeil Khoshnam
- Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Maryam Farzaneh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
33
|
Asgharzadeh F, Hashemzehi M, Moradi-Marjaneh R, Hassanian SM, Ferns GA, Khazaei M, Avan A. Angiotensin-converting enzyme inhibitors and angiotensin receptor blockers as therapeutic options in the treatment of renal cancer: A meta-analysis. Life Sci 2019; 242:117181. [PMID: 31863771 DOI: 10.1016/j.lfs.2019.117181] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/04/2019] [Accepted: 12/16/2019] [Indexed: 11/25/2022]
Abstract
AIMS Angiotensin-converting enzyme inhibitor (ACEI) and angiotensin receptor blocker (ARB) which have been used in the treatment of cardiovascular diseases, have also been shown to have anti-tumor effects against various cancers that include renal cancer. The aim of current paper was to explore the potential clinical impact of ACEI/ARB inhibitors in renal cancer. MAIN METHODS We used several databases: EMBASE, PubMed and the Cochrane library, to identify clinical studies that assessed the relationship between ACEIs/ARBs treatment and risk of renal cancer incidence or survival of renal cancer patients. The hazard ratio (HR) with 95% confidence intervals were obtained for assessing the relationship between ACEIs/ARBs and renal cancer mortality. KEY FINDING The HR for the relationship between ASIs use and survival of renal cancer indicated that patients with renal cancer being treated with ACEIs/ARBs had a significantly lower mortality than non-user (HR 0.723, 95% CI 0.568-0.921, p = 0.009). The HR for the relationship between ACEIs use and survival of renal cancer indicated that patients with renal cancer that used ACEIs had a higher mortality than non-users (HR 1.352, 95% CI 0.917-1.991, p = 0.128). The HR for the relationship between ARBs use and survival of renal cancer indicated that patients with renal cancer that used ARBs had a decreased of mortality than non-users (HR 0.818, 95% CI 0.691-0.969, p = 0.02). SIGNIFICANCE This meta-analysis demonstrated that treatment with ACEIs/ARBs may improve renal cancer survival and reduce the mortality of patients with renal cancer.
Collapse
Affiliation(s)
- Fereshteh Asgharzadeh
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Milad Hashemzehi
- Iranshahr University of Medical Sciences, Iranshahr, Iran; Department of Medical Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reyhaneh Moradi-Marjaneh
- Department of Physiology, School of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Seyed Mahdi Hassanian
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex BN1 9PH, UK
| | - Majid Khazaei
- Department of Medical Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Amir Avan
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
34
|
Abstract
The introduction of targeted agents into modern cancer therapy pursued the goal of molecularly more specific, and thereby more effective and safer, therapies. Paradoxically, however, several toxicities were brought to greater attention, among these not only cardiac but also vascular toxicities. The latter reach far beyond venous thromboembolism and include a broad spectrum of presentations based on the vascular territories and pathomechanisms involved, including abnormal vascular reactivity, acute thrombosis, or accelerated atherosclerosis. This article provides an overview of the most common presentations and their management strategies.
Collapse
Affiliation(s)
- Joerg Herrmann
- Department of Cardiovascular Diseases, Mayo Clinic, 200 First Street SW, Rochester, MN 55902, USA.
| |
Collapse
|
35
|
Dolomatov S, Zukow W, Novikov N, Markaryan A, Eremeeva E. EXPRESSION OF THE RENIN-ANGIOTENSIN SYSTEM COMPONENTS IN ONCOLOGIC DISEASES. Acta Clin Croat 2019; 58:354-364. [PMID: 31819334 PMCID: PMC6884393 DOI: 10.20471/acc.2019.58.02.21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The literature devoted to changes in the expression of the renin-angiotensin system (RAS) proteins of cancer cells was analyzed. The dynamics of RAS protein expression in malignant tumors and the possible role of epigenetic mechanisms in these processes are briefly reviewed. Through research of the epigenetic mechanisms in cancer, principally new techniques for their correction based on the use of selective regulatory systems of covalent modification of histone proteins (for example, deacetylase inhibitor) and microRNA synthesis technologies have been developed. Literature data show promising pharmacological correction of epigenetic modification of chromatin in the treatment of cancer.
Collapse
Affiliation(s)
| | - Walery Zukow
- 1Department of Medical Biology, Medical Academy SI Georgievsky, Crimea Federal University, Simferopol, Russian Federation jurisdiction; 2Faculty of Earth, Nicolaus Copernicus University, Toruń, Poland; 3A. Tsyb Medical Radiological Research Center, branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Kaluga Region, Russian Federation
| | - Nikolay Novikov
- 1Department of Medical Biology, Medical Academy SI Georgievsky, Crimea Federal University, Simferopol, Russian Federation jurisdiction; 2Faculty of Earth, Nicolaus Copernicus University, Toruń, Poland; 3A. Tsyb Medical Radiological Research Center, branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Kaluga Region, Russian Federation
| | - Alexandra Markaryan
- 1Department of Medical Biology, Medical Academy SI Georgievsky, Crimea Federal University, Simferopol, Russian Federation jurisdiction; 2Faculty of Earth, Nicolaus Copernicus University, Toruń, Poland; 3A. Tsyb Medical Radiological Research Center, branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Kaluga Region, Russian Federation
| | - Elena Eremeeva
- 1Department of Medical Biology, Medical Academy SI Georgievsky, Crimea Federal University, Simferopol, Russian Federation jurisdiction; 2Faculty of Earth, Nicolaus Copernicus University, Toruń, Poland; 3A. Tsyb Medical Radiological Research Center, branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Kaluga Region, Russian Federation
| |
Collapse
|
36
|
Alishahi M, Farzaneh M, Ghaedrahmati F, Nejabatdoust A, Sarkaki A, Khoshnam SE. NLRP3 inflammasome in ischemic stroke: As possible therapeutic target. Int J Stroke 2019; 14:574-591. [PMID: 30940045 DOI: 10.1177/1747493019841242] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Inflammation is a devastating pathophysiological process during stroke, a devastating disease that is the second most common cause of death worldwide. Activation of the NOD-like receptor protein (NLRP3)-infammasome has been proposed to mediate inflammatory responses during ischemic stroke. Briefly, NLRP3 inflammasome activates caspase-1, which cleaves both pro-IL-1 and pro-IL-18 into their active pro-inflammatory cytokines that are released into the extracellular environment. Several NLRP3 inflammasome inhibitors have been promoted, including small molecules, type I interferon, micro RNAs, nitric oxide, and nuclear factor erythroid-2 related factor 2 (Nrf2), some of which are potentially efficacious clinically. This review will describe the structure and cellular signaling pathways of the NLRP3 inflammasome during ischemic stroke, and current evidence for NLRP3 inflammasome inhibitors.
Collapse
Affiliation(s)
- Masoumeh Alishahi
- 1 Department of Biology, Tehran North Branch, Islamic Azad University, Tehran, Iran
| | - Maryam Farzaneh
- 2 Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Farhoodeh Ghaedrahmati
- 3 Immunology Department, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Armin Nejabatdoust
- 4 Department of Biology, Rasht Branch, Islamic Azad University, Rasht, Iran
| | - Alireza Sarkaki
- 5 Department of Physiology, Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyed Esmaeil Khoshnam
- 5 Department of Physiology, Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
37
|
Abstract
Nanotechnology offers new solutions for the development of cancer therapeutics that display improved efficacy and safety. Although several nanotherapeutics have received clinical approval, the most promising nanotechnology applications for patients still lie ahead. Nanoparticles display unique transport, biological, optical, magnetic, electronic, and thermal properties that are not apparent on the molecular or macroscale, and can be utilized for therapeutic purposes. These characteristics arise because nanoparticles are in the same size range as the wavelength of light and display large surface area to volume ratios. The large size of nanoparticles compared to conventional chemotherapeutic agents or biological macromolecule drugs also enables incorporation of several supportive components in addition to active pharmaceutical ingredients. These components can facilitate solubilization, protection from degradation, sustained release, immunoevasion, tissue penetration, imaging, targeting, and triggered activation. Nanoparticles are also processed differently in the body compared to conventional drugs. Specifically, nanoparticles display unique hemodynamic properties and biodistribution profiles. Notably, the interactions that occur at the bio-nano interface can be exploited for improved drug delivery. This review discusses successful clinically approved cancer nanodrugs as well as promising candidates in the pipeline. These nanotherapeutics are categorized according to whether they predominantly exploit multifunctionality, unique electromagnetic properties, or distinct transport characteristics in the body. Moreover, future directions in nanomedicine such as companion diagnostics, strategies for modifying the microenvironment, spatiotemporal nanoparticle transitions, and the use of extracellular vesicles for drug delivery are also explored.
Collapse
Affiliation(s)
- Joy Wolfram
- Department of Transplantation/Department of Physiology and Biomedical Engineering, Mayo Clinic, Jacksonville, Florida 32224, USA
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas 77030, USA
| | - Mauro Ferrari
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas 77030, USA
- Department of Medicine, Weill Cornell Medicine, Weill Cornell Medicine, New York, New York 10065, USA
| |
Collapse
|
38
|
Mozolevska V, Schwartz A, Cheung D, Goyal V, Shaikh B, Dingman B, Kim E, Mittal I, Asselin CY, Edel A, Ravandi A, Thliveris J, Singal PK, Czaykowski P, Jassal DS. Role of renin-angiotensin system antagonists in the prevention of bevacizumab- and sunitinib-mediated cardiac dysfunction. Am J Physiol Heart Circ Physiol 2019; 316:H446-H458. [DOI: 10.1152/ajpheart.00344.2018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Although anticancer systemic therapy agents clearly lead to improved survival in patients with cancer, these can come at the cost of serious complications including cardiotoxicity. Two types of targeted systemic therapies currently in use for colorectal cancer (CRC) and renal cell cancer (RCC), respectively, include the vascular endothelial growth factor inhibitor bevacizumab (BVZ) and the tyrosine kinase inhibitor sunitinib (SNT). Despite the beneficial effects of BVZ and SNT in improving clinical outcomes in the settings of CRC and RCC, there is an increased risk of cardiac dysfunction. The aim of the present study was to determine whether prophylactic administration of renin-angiotensin system (RAS) inhibitors would attenuate the cardiotoxic side effects of BVZ or SNT in a chronic in vivo murine model. A total of 194 wild-type C57Bl/6 male mice received: 1) 0.9% saline, 2) BVZ (10 mg·kg−1·wk−1), or 3) SNT (40 mg·kg−1·day−1) for 4 wk. Within each arm, mice received daily prophylactic treatment with hydralazine (0.05 mg/ml), aliskiren (50 mg/kg), perindopril (4 mg/kg), or valsartan (2 mg/kg). Although hydralazine effectively lowered blood pressure in BVZ- or SNT-treated mice, it did not prevent left ventricular systolic dysfunction. Prophylactic administration of aliskiren, perindopril, or valsartan prevented adverse cardiovascular remodeling in mice treated with either BVZ or SNT. The addition of RAS antagonists also downregulated expression of phosphorylated p38 and Bcl-2-like 19-kDa interacting protein 3 in SNT-treated mice. In our chronic in vivo murine model, RAS antagonists partially attenuated the development of BVZ- or SNT-mediated cardiac dysfunction. Future clinical studies are warranted to investigate the cardioprotective effects of prophylactic treatment with RAS inhibitors in the settings of CRC and RCC. NEW & NOTEWORTHY In the evolving field of cardio-oncology, bevacizumab and sunitinib improve clinical outcomes in the settings of metastatic colorectal cancer and renal cell cancer, respectively. These anticancer drugs, however, are associated with an increased risk of cardiotoxicity. The prophylactic administration of renin-angiotensin system antagonists is partially cardioprotective against bevacizumab- and sunitinib-mediated cardiac dysfunction.
Collapse
Affiliation(s)
- Viktoriya Mozolevska
- Institute of Cardiovascular Sciences, Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Anna Schwartz
- Institute of Cardiovascular Sciences, Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - David Cheung
- Institute of Cardiovascular Sciences, Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Vineet Goyal
- Institute of Cardiovascular Sciences, Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Bilal Shaikh
- Institute of Cardiovascular Sciences, Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Bella Dingman
- Institute of Cardiovascular Sciences, Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Esther Kim
- Section of Cardiology, Department of Internal Medicine, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Ishika Mittal
- Institute of Cardiovascular Sciences, Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Chantal Y. Asselin
- Institute of Cardiovascular Sciences, Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Andrea Edel
- Institute of Cardiovascular Sciences, Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Amir Ravandi
- Institute of Cardiovascular Sciences, Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
- Section of Cardiology, Department of Internal Medicine, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - James Thliveris
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Pawan K. Singal
- Institute of Cardiovascular Sciences, Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Piotr Czaykowski
- Section of Hematology/Oncology, Department of Internal Medicine, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Davinder S. Jassal
- Institute of Cardiovascular Sciences, Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
- Section of Cardiology, Department of Internal Medicine, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
- Section of Hematology/Oncology, Department of Internal Medicine, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Radiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
39
|
Johansen MD, Urup T, Holst CB, Christensen IJ, Grunnet K, Lassen U, Friis S, Poulsen HS. Outcome of Bevacizumab Therapy in Patients with Recurrent Glioblastoma Treated with Angiotensin System Inhibitors. Cancer Invest 2019; 36:512-519. [PMID: 30727779 DOI: 10.1080/07357907.2018.1544639] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
PURPOSE Antihypertensive therapy may improve bevacizumab efficacy in cancer patients. We examined efficacy and toxicity of angiotensin system inhibitors (ASI) and other antihypertensive drugs in bevacizumab treated recurrent glioblastoma patients. METHODS We retrospectively combined a national prescription registry with a clinical database with recurrent glioblastoma patients (n = 243). RESULTS Patients who initiated ASI after bevacizumab (n = 26) showed a tendency towards improved progression-free survival and overall survival (OS) with hazard rate (HR) reductions (HR = 0.70 and HR = 0.79, respectively). Calcium antagonists during bevacizumab therapy significantly improved OS (HR = 0.57). CONCLUSIONS Overall the study supports a potential beneficial effect of antihypertensive treatment on prognosis of bevacizumab treated glioblastoma patients.
Collapse
Affiliation(s)
- Maria Dinche Johansen
- a Department of Radiation Biology, Department of Oncology , The Finsen Center , Rigshospitalet, Copenhagen , Denmark
| | - Thomas Urup
- a Department of Radiation Biology, Department of Oncology , The Finsen Center , Rigshospitalet, Copenhagen , Denmark
| | - Camilla Bjørnbak Holst
- a Department of Radiation Biology, Department of Oncology , The Finsen Center , Rigshospitalet, Copenhagen , Denmark
| | | | - Kirsten Grunnet
- a Department of Radiation Biology, Department of Oncology , The Finsen Center , Rigshospitalet, Copenhagen , Denmark
| | - Ulrik Lassen
- c Department of Oncology , The Finsen Center , Rigshospitalet, Copenhagen , Denmark
| | - Søren Friis
- d Danish Cancer Society , Danish Cancer Society Research Center , Copenhagen , Denmark
| | - Hans Skovgaard Poulsen
- a Department of Radiation Biology, Department of Oncology , The Finsen Center , Rigshospitalet, Copenhagen , Denmark
| |
Collapse
|
40
|
Ursu R, Thomas L, Psimaras D, Chinot O, Le Rhun E, Ricard D, Charissoux M, Cuzzubbo S, Sejalon F, Quillien V, Hoang-Xuan K, Ducray F, Portal JJ, Tibi A, Mandonnet E, Levy-Piedbois C, Vicaut E, Carpentier AF. Angiotensin II receptor blockers, steroids and radiotherapy in glioblastoma-a randomised multicentre trial (ASTER trial). An ANOCEF study. Eur J Cancer 2019; 109:129-136. [PMID: 30716716 DOI: 10.1016/j.ejca.2018.12.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 12/18/2018] [Accepted: 12/23/2018] [Indexed: 11/20/2022]
Abstract
BACKGROUND Glioblastomas (GBMs) induce a peritumoural vasogenic oedema impairing functional status and quality of life. Steroids reduce brain tumour-related oedema but are associated with numerous side-effects. It was reported in a retrospective series that angiotensin receptor blockers might be associated with reduced peritumoural oedema. The ASTER study is a randomised, placebo-controlled trial to assess whether or not the addition of Losartan to standard of care (SOC) can reduce steroid requirement during radiotherapy (RT) in patients with newly diagnosed GBM. PATIENTS AND METHODS Patients with a histologically confirmed GBM after biopsy or partial surgical resection were randomised between Losartan or placebo in addition to SOC with RT and temozolomide (TMZ). The primary objective was to investigate the steroid dosage required to control brain oedema on the last day of RT in each arm. The secondary outcomes were steroids dosage 1 month after the end of RT, assessment of cerebral oedema on magnetic resonance imaging, tolerance and survival. RESULTS Seventy-five patients were randomly assigned to receive Losartan (37 patients) or placebo (38 patients). No difference in the steroid dosage required to control brain oedema on the last day of RT, or one month after completion of RT, was seen between both arms. The incidence of adverse events was similar in both arms. Median overall survival was similar in both arms. CONCLUSIONS Losartan, although well tolerated, does not reduce the steroid requirement in newly diagnosed GBM patients treated with concomitant RT and TMZ. Trial registration number NCT01805453 with ClinicalTrials.gov.
Collapse
Affiliation(s)
- R Ursu
- Department of Neurology, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France; University Paris Diderot, Sorbonne Paris Cité, Paris, France.
| | - L Thomas
- Department of Neuro-Oncology, Hospices Civils de Lyon, Groupe Hospitalier Est, Lyon, France
| | - D Psimaras
- Department of Neurology Mazarin, Assistance Publique-Hôpitaux de Paris, Sorbonne Université, Hôpital Pitié-Salpêtrière, Paris, France
| | - O Chinot
- Department of Neuro-Oncology, CHU Timone, Marseille, France; Aix-Marseille Université, Assistance Publique-Hôpitaux de Marseille, Marseille, France
| | - E Le Rhun
- University of Lille, Inserm, U-1192, F-59000 Lille, France; CHU Lille, General and Stereotaxic Neurosurgery Service, F-59000 Lille, France; Oscar Lambret Center, Neurology, Medical Oncology Department, F-59000 Lille, France
| | - D Ricard
- Department of Neurology, Hôpital d'Instruction des Armées Percy, Service de Santé des Armées, Paris, France
| | - M Charissoux
- Department of Radiation Oncology, Institut du Cancer de Montpellier, Montpellier cedex 5, France
| | - S Cuzzubbo
- Department of Neurology, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France; University Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - F Sejalon
- Department of Neurology, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - V Quillien
- Centre de Lutte Contre le Cancer Eugène Marquis, F-35042 Rennes, France; INSERM U1242, "Chemistry, Oncogenesis, Stress, Signaling", Université de Rennes 1, Rennes, France
| | - K Hoang-Xuan
- Department of Neurology Mazarin, Assistance Publique-Hôpitaux de Paris, Sorbonne Université, Hôpital Pitié-Salpêtrière, Paris, France
| | - F Ducray
- Department of Neuro-Oncology, Hospices Civils de Lyon, Groupe Hospitalier Est, Lyon, France; Department of Cancer Cell Plasticity, Cancer Research Centre of Lyon, INSERM U1052, CNRS UMR5286, Lyon, France; University Claude Bernard Lyon 1, Lyon, France
| | - J-J Portal
- AP-HP, Unité de Recherche Clinique, Hôpital Fernand Widal, Université Paris-Diderot, Paris, France
| | - A Tibi
- Agence Générale des Equipements et Produits de Santé (AGEPS), Paris, France
| | - E Mandonnet
- Department of Neurosurgery, Lariboisière Hospital, APHP, Paris, France; University Paris 7, Paris, France; IMNC, UMR 8165, Orsay, France
| | - C Levy-Piedbois
- Ramsey Générale de Santé, Institut de Radiothérapie des Hauts-Energies, Bobigny, France
| | - E Vicaut
- AP-HP, Unité de Recherche Clinique, Hôpital Fernand Widal, Université Paris-Diderot, Paris, France
| | - A F Carpentier
- Department of Neurology, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France; University Paris Diderot, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
41
|
The De Ritis and Neutrophil-to-Lymphocyte Ratios May Aid in the Risk Assessment of Patients with Metastatic Renal Cell Carcinoma. JOURNAL OF ONCOLOGY 2018; 2018:1953571. [PMID: 30662462 PMCID: PMC6312581 DOI: 10.1155/2018/1953571] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 10/14/2018] [Indexed: 01/05/2023]
Abstract
Purpose This study aimed to determine whether baseline blood inflammatory markers can predict progression-free survival (PFS) and overall survival (OS) in patients with metastatic renal cell carcinoma (mRCC). Methods The study included 158 patients with mRCC treated with first-line targeted therapy between 2002 and 2016. A multivariable cox proportional hazards model identified inflammatory factors that predict PFS and OS. Using bootstrap method, new prognostic model compared with Heng and modified MSKCC risk model (mMSKCC). The effect of inflammatory factors were investigated by comparing increased C-index adding significant inflammatory factors to Heng and mMSKCC model. Results On multivariable analysis, nephrectomy (HR 0.48), NLR (HR 1.04), were significant risk factors for PFS; nephrectomy (HR 0.38), hemoglobin (HR 1.71), alkaline phosphatase (HR 1.73), NLR (HR 1.01) and DRR (HR 1.34), were significant factors for OS (p<0.05). Our new model that incorporated NLR and DRR had higher (though insignificant) predictability (C-index=0.610) than mMSKCC risk model (C-index=0.569) in PFS and significantly better predictability (C-index=0.727) than Heng and mMSKCC risk model (C-index, 0.661, 0.612, respectively) in OS. Adding inflammatory factors to the Heng criteria (C-index, 0.697 for OS) and MSKCC (0.691 for OS) tended to improve their predictive abilities. Conclusions The NLR and DRR may increase predictive ability compared to the established Heng and mMSKCC risk models in mRCC.
Collapse
|
42
|
Vallejo-Ardila DL, Fifis T, Burrell LM, Walsh K, Christophi C. Renin-angiotensin inhibitors reprogram tumor immune microenvironment: A comprehensive view of the influences on anti-tumor immunity. Oncotarget 2018; 9:35500-35511. [PMID: 30464806 PMCID: PMC6231452 DOI: 10.18632/oncotarget.26174] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 09/08/2018] [Indexed: 12/30/2022] Open
Abstract
Renin-angiotensin system inhibitors (RASi) have shown potential anti-tumor effects that may have a significant impact in cancer therapy. The components of the renin-angiotensin system (RAS) including both, conventional and alternative axis, appear to have contradictory effects on tumor biology. The mechanisms by which RASi impair tumor growth extend beyond their function of modulating tumor vasculature. The major focus of this review is to analyze other mechanisms by which RASi reprogram the tumor immune microenvironment. These involve impairing hypoxia and acidosis within the tumor stroma, regulating inflammatory signaling pathways and oxidative stress, modulating the function of the non-cellular components and immune cells, and regulating the cross-talk between kalli krein kinin system and RAS.
Collapse
Affiliation(s)
- Dora L Vallejo-Ardila
- Department of Surgery, Austin Health, University of Melbourne, Melbourne,VIC 3084, Australia
| | - Theodora Fifis
- Department of Surgery, Austin Health, University of Melbourne, Melbourne,VIC 3084, Australia
| | - Louise M Burrell
- Department of Medicine, Austin Health, University of Melbourne, Melbourne, VIC 3084, Australia.,Department of Cardiology, Austin Health, University of Melbourne, Melbourne, VIC 3084, Australia
| | - Katrina Walsh
- Department of Surgery, Austin Health, University of Melbourne, Melbourne,VIC 3084, Australia
| | - Christopher Christophi
- Department of Surgery, Austin Health, University of Melbourne, Melbourne,VIC 3084, Australia
| |
Collapse
|
43
|
Gelfond J, Al-Bayati O, Kabra A, Iffrig K, Kaushik D, Liss MA. Modifiable risk factors to reduce renal cell carcinoma incidence: Insight from the PLCO trial. Urol Oncol 2018; 36:340.e1-340.e6. [PMID: 29779672 DOI: 10.1016/j.urolonc.2018.04.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 03/07/2018] [Accepted: 04/17/2018] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Identify modifiable factors contributing to renal cell carcinoma in the PCLO to target disease prevention and reduce health care costs. METHODS The prostate, lung, colorectal, and ovarian database were queried for the primary outcome of kidney cancer. Demographics were investigated, specifically focusing on modifiable risk factors. Statistical analysis includes the Student t-test for continuous variables, chi-squared or Fisher's exact tests for dichotomous and categorical variables for bivariate analysis. The Cox proportional hazards model was used in a multivariate time-to-event analysis. RESULTS We investigate existing data relating specifically to renal cancer. After missing data were excluded, we analyzed 149,683 subjects enrolled in the prostate, lung, colorectal, and ovarian trial and noted 0.5% (n = 748) subjects developed renal cancer. Age, male gender, body mass index, diabetes, and hypertension were all significant associated with renal cancer in bivariate analysis (P<0.05). Men have a significant increased risk of kidney cancer over women (hazard ratio [HR] = 1.85; 95% CI: 1.58-2.16; P<0.0001). Nonmodifiable risk factors that are associated with kidney cancer include age (HR = 1.05; 95% CI: 1.01; 1.05, P = 0.001). Modifiable risk factors include obesity measured by body mass index (HR = 1.05; 95% CI: 1.02-1.07; P<0.0001), hypertension (HR = 1.32; 95% CI: 1.13-1.54; P = 0.0004), and smoking in pack-years (HR = 1.04; 95% CI: 1.02-1.07; P = 0.0002). CONCLUSIONS Obesity, hypertension, and smoking are the 3 modifiable risk factors that could aggressively be targeted to reduce renal cell carcinoma.
Collapse
Affiliation(s)
- Jonathan Gelfond
- Department of Biostatistics, University of Texas Health Science Center San Antonio, San Antonio, TX
| | - Osamah Al-Bayati
- Department of Urology, University of Texas Health Science Center San Antonio, San Antonio, TX
| | - Aashish Kabra
- Department of Urology, University of Texas Health Science Center San Antonio, San Antonio, TX
| | - Kevan Iffrig
- Department of Urology, University of Texas Health Science Center San Antonio, San Antonio, TX
| | - Dharam Kaushik
- Department of Urology, University of Texas Health Science Center San Antonio, San Antonio, TX
| | - Michael A Liss
- Department of Urology, University of Texas Health Science Center San Antonio, San Antonio, TX; Department of Surgery, South Texas Veterans Healthcare System, San Antonio, TX.
| |
Collapse
|
44
|
Caletti S, Paini A, Coschignano MA, De Ciuceis C, Nardin M, Zulli R, Muiesan ML, Salvetti M, Rizzoni D. Management of VEGF-Targeted Therapy-Induced Hypertension. Curr Hypertens Rep 2018; 20:68. [PMID: 29959593 DOI: 10.1007/s11906-018-0871-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW From a physiological point of view, VEGFs (vascular endothelial growth factors) and their receptors (VEGFR) play a critical role in vascular development angiogenesis, endothelial function, and vascular tone. On the pathological side, VEGF-VEGFR signaling may induce dysregulated angiogenesis, which contributes to the growth and to the spread of tumors, being essential for neoplastic proliferation and invasion. RECENT FINDINGS Pharmacological inhibition of VEGF-VEGFR is now a cornerstone in the treatment of many malignancies; however, treatment with VEGF inhibitors is commonly associated with an increase in blood pressure values. This side effect is strictly connected with the mechanism of action of these medications and might represent an index of therapy efficacy. The optimal management of this form of hypertension is, at present, not clear. Calcium channel blockers and renin-angiotensin system inhibitors probably represent the most appropriate classes of hypertensive dugs for the treatment of this condition; however, no conclusive data are presently available.
Collapse
Affiliation(s)
- Stefano Caletti
- Clinica Medica, Department of Medical and Surgical Sciences, University of Brescia, c/o 2a Medicina Spedali Civili di Brescia, Piazza Spedali Civili 1, 25100, Brescia, Italy
| | - Anna Paini
- Clinica Medica, Department of Medical and Surgical Sciences, University of Brescia, c/o 2a Medicina Spedali Civili di Brescia, Piazza Spedali Civili 1, 25100, Brescia, Italy
| | - Maria Antonietta Coschignano
- Clinica Medica, Department of Medical and Surgical Sciences, University of Brescia, c/o 2a Medicina Spedali Civili di Brescia, Piazza Spedali Civili 1, 25100, Brescia, Italy
| | - Carolina De Ciuceis
- Clinica Medica, Department of Medical and Surgical Sciences, University of Brescia, c/o 2a Medicina Spedali Civili di Brescia, Piazza Spedali Civili 1, 25100, Brescia, Italy
| | - Matteo Nardin
- Clinica Medica, Department of Medical and Surgical Sciences, University of Brescia, c/o 2a Medicina Spedali Civili di Brescia, Piazza Spedali Civili 1, 25100, Brescia, Italy
| | - Roberto Zulli
- Clinica Medica, Department of Medical and Surgical Sciences, University of Brescia, c/o 2a Medicina Spedali Civili di Brescia, Piazza Spedali Civili 1, 25100, Brescia, Italy
| | - Maria Lorenza Muiesan
- Clinica Medica, Department of Medical and Surgical Sciences, University of Brescia, c/o 2a Medicina Spedali Civili di Brescia, Piazza Spedali Civili 1, 25100, Brescia, Italy
| | - Massimo Salvetti
- Clinica Medica, Department of Medical and Surgical Sciences, University of Brescia, c/o 2a Medicina Spedali Civili di Brescia, Piazza Spedali Civili 1, 25100, Brescia, Italy
| | - Damiano Rizzoni
- Clinica Medica, Department of Medical and Surgical Sciences, University of Brescia, c/o 2a Medicina Spedali Civili di Brescia, Piazza Spedali Civili 1, 25100, Brescia, Italy.
| |
Collapse
|
45
|
Saber S, Mahmoud A, Helal N, El-Ahwany E, Abdelghany R. Liver Protective Effects of Renin-Angiotensin System Inhibition Have No Survival Benefits in Hepatocellular Carcinoma Induced By Repetitive Administration of Diethylnitrosamine in Mice. Open Access Maced J Med Sci 2018; 6:955-960. [PMID: 29983784 PMCID: PMC6026411 DOI: 10.3889/oamjms.2018.167] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 03/17/2018] [Accepted: 03/23/2018] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND: Preclinical studies have demonstrated that renin-angiotensin system (RAS) signalling has strong tumour-promoting effects and RAS inhibition was associated with improvement in the overall survival in some cancer types including hepatocellular carcinoma (HCC). OBJECTIVE: We aimed to investigate the effect of angiotensin-converting enzyme inhibitors (ACEIs) or angiotensin-II-receptor blockers (ARBs) on the survival of mice with diethylnitrosamine (DEN) induced HCC. METHODS: HCC was induced by weekly i.p. administration of DEN. Mice were treated with sorafenib (SO) (30 mg/kg), perindopril (PE) (1 mg/kg), fosinopril (FO) (2 mg/kg), losartan (LO) (10 mg/kg), PE (1 mg/kg) + SO (30 mg/kg), FO (2 mg/kg) + SO (30 mg/kg), or LO (10 mg/kg) + SO (30 mg/kg). Survival analysis was done using the Kaplan-Meier method, and the log-rank test was used for assessing the significance of difference between groups. RESULTS: The administration of PE, FO and LO as monotherapy or as combined with SO resulted in marked improvement in the liver histologic picture with no impact on overall survival of mice. CONCLUSION: Interfering the RAS either through the inhibition of ACE or the blockade of angiotensin II type 1 (AT1) receptors has similar effects on the liver of DEN-induced HCC mice and is not associated with longer survival due to detrimental effects of DEN on other organs. Hence, repetitive administration of DEN in such models of HCC is not suitable for mortality assessment studies.
Collapse
Affiliation(s)
- Sameh Saber
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| | - Amr Mahmoud
- Department of Pharmacology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Noha Helal
- Pathology Department, Theodor Bilharz Research Institute, Giza, Egypt
| | - Eman El-Ahwany
- Immunology Department, Theodor Bilharz Research Institute, Giza, Egypt
| | - Rasha Abdelghany
- Department of Pharmacology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| |
Collapse
|
46
|
Pinter M, Jain RK. Targeting the renin-angiotensin system to improve cancer treatment: Implications for immunotherapy. Sci Transl Med 2018; 9:9/410/eaan5616. [PMID: 28978752 PMCID: PMC5928511 DOI: 10.1126/scitranslmed.aan5616] [Citation(s) in RCA: 214] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 08/25/2017] [Indexed: 12/25/2022]
Abstract
Renin-angiotensin system (RAS) inhibitors (RASi)-widely prescribed for the treatment of cardiovascular diseases-have considerable potential in oncology. The RAS plays a crucial role in cancer biology and affects tumor growth and dissemination directly and indirectly by remodeling the tumor microenvironment. We review clinical data on the benefit of RASi in primary and metastatic tumors and propose that, by activating immunostimulatory pathways, these inhibitors can enhance immunotherapy of cancer.
Collapse
Affiliation(s)
- Matthias Pinter
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Harvard Medical School and Massachusetts General Hospital, Boston, MA 02114, USA.,Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, A-1090, Austria
| | - Rakesh K Jain
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Harvard Medical School and Massachusetts General Hospital, Boston, MA 02114, USA.
| |
Collapse
|
47
|
Touyz RM, Herrmann SMS, Herrmann J. Vascular toxicities with VEGF inhibitor therapies-focus on hypertension and arterial thrombotic events. ACTA ACUST UNITED AC 2018; 12:409-425. [PMID: 29703600 PMCID: PMC6168784 DOI: 10.1016/j.jash.2018.03.008] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 03/02/2018] [Accepted: 03/14/2018] [Indexed: 12/21/2022]
Abstract
The vascular endothelial growth factor (VEGF) signaling pathway (VSP) fulfills a cardinal role in endothelial cells and its inhibition has profound cardiovascular impact. This is true not only for the normal vasculature but also for the tumor vasculature when VSP inhibitors are used as anti-angiogenic therapies. Generalized endothelial dysfunction predisposes to vasoconstriction, atherosclerosis, platelet activation, and thrombosis (arterial more than venous). All of these have been reported with VSP inhibitors and collectively give rise to vascular toxicities, the most concerning of which are arterial thromboembolic events (ATE). VSP inhibitors include antibodies, acting extracelluarly on VEGF, such as bevacizumab and tyrosine kinases inhibitors, acting intracellularly on the kinase domain of VEGF receptors, such as sunintib and sorafenib. The addition of bevacizumab and VSP tyrosine kinase inhibitor therapy to the cancer treatment regimen is associated with a 1.5-2.5-fold and 2.3-4.6-fold increase risk of ATEs, respectively. Risk factors for ATEs while on VSP inhibitor therapy include age older than 65 years, previous thromboembolic events, history of atherosclerotic disease, and duration of VSP inhibitor therapy. In clinical practice, hypertension remains the most commonly noted vascular manifestation of VSP inhibition. Optimal blood pressure goals and preferred therapeutic strategies toward reaching these goals are not defined at present. This review summarizes current data on this topic and proposes a more intensive management approach to patients undergoing VSP inhibitor therapy including Systolic Blood PRessure Intervention Trial (SPRINT) blood pressure goals, pleiotropic vasoprotective agents such as angiotensin converting enzyme inhibitors, amlodipine, and carvedilol, high-dose statin therapy, and aspirin.
Collapse
Affiliation(s)
- Rhian M Touyz
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Sandra M S Herrmann
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Joerg Herrmann
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
48
|
The effect of angiotensin system inhibitors (angiotensin-converting enzyme inhibitors or angiotensin receptor blockers) on cancer recurrence and survival: a meta-analysis. Eur J Cancer Prev 2018; 26:78-85. [PMID: 27158979 DOI: 10.1097/cej.0000000000000269] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
To assess the current evidence on the potential benefit of angiotensin-converting enzyme inhibitors (ACEIs) or angiotensin receptor blockers (ARBs) on cancer recurrence and survival, we comprehensively searched PubMed, Embase, and the Cochrane Library from their inception to April 2013. Two authors screened out duplicates and independently reviewed the eligibility of each study. We included comparative studies comparing the use and nonuse of ACEIs or ARBs in cancer patients. Primary outcomes were disease-free survival (DFS) and overall survival. We included 11 studies with 4964 participants in the final analysis. The meta-analysis showed that the use of ACEIs or ARBs resulted in a significant improvement in DFS [hazard ratio (HR) 0.60; 95% confidence interval (CI) 0.41-0.87; P=0.007)] and overall survival (HR 0.75; 95% CI 0.57-0.99; P=0.04). Even when cancer stage was classified into low (I/II) or high (III/IV), DFS improvement was applied to both low stage (HR 0.56; 95% CI 0.32-0.96; P=0.04) and high stage (HR 0.59; 95% CI 0.37-0.94; P=0.03). Analysis according to cancer type showed benefits in urinary tract cancer (HR 0.22), colorectal cancer (HR 0.22), pancreatic cancer (HR 0.58), and prostate cancer (HR 0.14), but not in breast cancer and hepatocellular cancer. This meta-analysis provides evidence that the use of ACEIs or ARBs in cancer patients can lead to a 40 and 25% reduction in the risk of cancer recurrence and mortality.
Collapse
|
49
|
Coulson R, Liew SH, Connelly AA, Yee NS, Deb S, Kumar B, Vargas AC, O'Toole SA, Parslow AC, Poh A, Putoczki T, Morrow RJ, Alorro M, Lazarus KA, Yeap EFW, Walton KL, Harrison CA, Hannan NJ, George AJ, Clyne CD, Ernst M, Allen AM, Chand AL. The angiotensin receptor blocker, Losartan, inhibits mammary tumor development and progression to invasive carcinoma. Oncotarget 2017; 8:18640-18656. [PMID: 28416734 PMCID: PMC5386636 DOI: 10.18632/oncotarget.15553] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 02/07/2017] [Indexed: 01/06/2023] Open
Abstract
Drugs that target the Renin-Angiotensin System (RAS) have recently come into focus for their potential utility as cancer treatments. The use of Angiotensin Receptor Blockers (ARBs) and Angiotensin-Converting Enzyme (ACE) Inhibitors (ACEIs) to manage hypertension in cancer patients is correlated with improved survival outcomes for renal, prostate, breast and small cell lung cancer. Previous studies demonstrate that the Angiotensin Receptor Type I (AT1R) is linked to breast cancer pathogenesis, with unbiased analysis of gene-expression studies identifying significant up-regulation of AGTR1, the gene encoding AT1R in ER+ve/HER2−ve tumors correlating with poor prognosis. However, there is no evidence, so far, of the functional contribution of AT1R to breast tumorigenesis. We explored the potential therapeutic benefit of ARB in a carcinogen-induced mouse model of breast cancer and clarified the mechanisms associated with its success. Mammary tumors were induced with 7,12-dimethylbenz[α]antracene (DMBA) and medroxyprogesterone acetate (MPA) in female wild type mice and the effects of the ARB, Losartan treatment assessed in a preventative setting (n = 15 per group). Tumor histopathology was characterised by immunohistochemistry, real-time qPCR to detect gene expression signatures, and tumor cytokine levels measured with quantitative bioplex assays. AT1R was detected with radiolabelled ligand binding assays in fresh frozen tumor samples. We showed that therapeutic inhibition of AT1R, with Losartan, resulted in a significant reduction in tumor burden; and no mammary tumor incidence in 20% of animals. We observed a significant reduction in tumor progression from DCIS to invasive cancer with Losartan treatment. This was associated with reduced tumor cell proliferation and a significant reduction in IL-6, pSTAT3 and TNFα levels. Analysis of tumor immune cell infiltrates, however, demonstrated no significant differences in the recruitment of lymphocytes or tumour-associated macrophages in Losartan or vehicle-treated mammary tumors. Analysis of AT1R expression with radiolabelled ligand binding assays in human breast cancer biopsies showed high AT1R levels in 30% of invasive ductal carcinomas analysed. Furthermore, analysis of the TCGA database identified that high AT1R expression to be associated with luminal breast cancer subtype. Our in vivo data and analysis of human invasive ductal carcinoma samples identify the AT1R is a potential therapeutic target in breast cancer, with the availability of a range of well-tolerated inhibitors currently used in clinics. We describe a novel signalling pathway critical in breast tumorigenesis, that may provide new therapeutic avenues to complement current treatments.
Collapse
Affiliation(s)
- Rhiannon Coulson
- Cancer Drug Discovery, Hudson's Institute of Medical Research, Clayton, VIC, Australia.,Translational Breast Cancer Research, Garvan Institute, Darlinghurst, Sydney, NSW, Australia
| | - Seng H Liew
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | | | - Nicholas S Yee
- Cancer and Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
| | - Siddhartha Deb
- Anatomical Pathology, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
| | - Beena Kumar
- Anatomical Pathology, Monash Health, Clayton, VIC, Australia
| | - Ana C Vargas
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, NSW, Australia
| | - Sandra A O'Toole
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, NSW, Australia.,Translational Breast Cancer Research, Garvan Institute, Darlinghurst, Sydney, NSW, Australia.,Sydney Medical School, Sydney University, NSW, Australia
| | - Adam C Parslow
- Tumor Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia.,School of Cancer Medicine, La Trobe University, Heidelberg, VIC, Australia
| | - Ashleigh Poh
- Inflammation Division, Walter and Eliza Hall Institute of Medical Research, VIC, Australia
| | - Tracy Putoczki
- Inflammation Division, Walter and Eliza Hall Institute of Medical Research, VIC, Australia
| | - Riley J Morrow
- Cancer and Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
| | - Mariah Alorro
- Cancer and Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
| | - Kyren A Lazarus
- Cancer Drug Discovery, Hudson's Institute of Medical Research, Clayton, VIC, Australia.,Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Evie F W Yeap
- Cancer Drug Discovery, Hudson's Institute of Medical Research, Clayton, VIC, Australia
| | - Kelly L Walton
- Department of Physiology, Monash University, Clayton, VIC, Australia
| | - Craig A Harrison
- Department of Physiology, Monash University, Clayton, VIC, Australia
| | - Natalie J Hannan
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, University of Melbourne, Mercy Hospital, Heidelberg, VIC, Australia
| | - Amee J George
- The ACRF Department of Cancer Biology and Therapeutics, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Colin D Clyne
- Cancer Drug Discovery, Hudson's Institute of Medical Research, Clayton, VIC, Australia
| | - Matthias Ernst
- Cancer and Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia.,School of Cancer Medicine, La Trobe University, Heidelberg, VIC, Australia
| | - Andrew M Allen
- Department of Physiology, University of Melbourne, VIC, Australia
| | - Ashwini L Chand
- Cancer Drug Discovery, Hudson's Institute of Medical Research, Clayton, VIC, Australia.,Cancer and Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia.,School of Cancer Medicine, La Trobe University, Heidelberg, VIC, Australia
| |
Collapse
|
50
|
Pinter M, Weinmann A, Wörns MA, Hucke F, Bota S, Marquardt JU, Duda DG, Jain RK, Galle PR, Trauner M, Peck-Radosavljevic M, Sieghart W. Use of inhibitors of the renin-angiotensin system is associated with longer survival in patients with hepatocellular carcinoma. United European Gastroenterol J 2017; 5:987-996. [PMID: 29163965 PMCID: PMC5676550 DOI: 10.1177/2050640617695698] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 02/01/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Inhibition of the renin-angiotensin system (RAS) was associated with longer survival in patients with different solid malignancies. OBJECTIVE The objective of this study was to investigate the effect of RAS inhibitor (RASi) treatment (angiotensin-converting enzyme inhibitors or angiotensin-II-receptor blockers) on survival of patients with hepatocellular carcinoma (HCC). METHODS Patients diagnosed with HCC and Child-Pugh A between 1992 and 2013 who received sorafenib, experimental therapy, or best supportive care were eligible for the Vienna cohort. The Mainz cohort included patients with HCC and Child-Pugh A who received sorafenib treatment between 2007 and 2016. The association between RASi and overall survival (OS) was evaluated in univariate and multivariate analyses. RESULTS In the Vienna cohort, 43 of 156 patients received RASi for hypertension. RASi treatment was associated with longer OS (11.9 vs. 6.8 months (mo); p = 0.014) and remained a significant prognostic factor upon multivariate analysis (HR = 0.6; 95% CI 0.4-0.9; p = 0.011). In subgroup analysis, patients treated with sorafenib plus RASi had better median OS (19.5 mo) compared to those treated with either sorafenib (10.9 mo) or RASi (9.7 mo) alone (p = 0.043). The beneficial effect of RASi on survival was confirmed in the Mainz cohort (n = 76). CONCLUSION RAS inhibition is associated with longer survival in HCC patients with Child-Pugh class A.
Collapse
Affiliation(s)
- Matthias Pinter
- Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
- Department of Radiation Oncology, Harvard Medical School & Massachusetts General Hospital, Boston, MA, USA
| | - Arndt Weinmann
- Department of Medicine I, University Medical Center Johannes Gutenberg University, Mainz, Germany
- Cirrhosis Center Mainz (CCM), University Medical Center Johannes Gutenberg University, Mainz, Germany
- Clinical Registry Unit (CRU), University Medical Center Johannes Gutenberg University, Mainz, Germany
| | - Marcus-Alexander Wörns
- Department of Medicine I, University Medical Center Johannes Gutenberg University, Mainz, Germany
- Cirrhosis Center Mainz (CCM), University Medical Center Johannes Gutenberg University, Mainz, Germany
| | - Florian Hucke
- Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
- Department of Gastroenterology & Hepatology, Endocrinology and Nephrology, Klinikum Klagenfurt am Wörthersee, Klagenfurt, Austria
| | - Simona Bota
- Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
- Department of Gastroenterology & Hepatology, Endocrinology and Nephrology, Klinikum Klagenfurt am Wörthersee, Klagenfurt, Austria
| | - Jens U Marquardt
- Department of Medicine I, University Medical Center Johannes Gutenberg University, Mainz, Germany
- Cirrhosis Center Mainz (CCM), University Medical Center Johannes Gutenberg University, Mainz, Germany
| | - Dan G Duda
- Department of Radiation Oncology, Harvard Medical School & Massachusetts General Hospital, Boston, MA, USA
| | - Rakesh K Jain
- Department of Radiation Oncology, Harvard Medical School & Massachusetts General Hospital, Boston, MA, USA
| | - Peter R Galle
- Department of Medicine I, University Medical Center Johannes Gutenberg University, Mainz, Germany
- Cirrhosis Center Mainz (CCM), University Medical Center Johannes Gutenberg University, Mainz, Germany
| | - Michael Trauner
- Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Markus Peck-Radosavljevic
- Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
- Department of Gastroenterology & Hepatology, Endocrinology and Nephrology, Klinikum Klagenfurt am Wörthersee, Klagenfurt, Austria
| | - Wolfgang Sieghart
- Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| |
Collapse
|